Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Glycobiology ; 33(4): 325-341, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-36790131

RESUMEN

In Parkinson's disease, there is an accumulation of α-synuclein (SYN) aggregates in neurons, which is promoted by neuroinflammation. In neural cells, cytokine-induced SYN aggregation is modulated by heparan sulfate (HS) derived from glypican-1 (GPC1) by amyloid precursor protein (APP) and nitric oxide (NO)-dependent cleavage. We have explored possible interplay between APP, GPC1, and SYN in undifferentiated and differentiated neural progenitor cells (NPCs) by modulating APP and GPC1 processing. Effects were monitored by immunofluorescence microscopy and slot immunoblotting using antibodies recognizing APP degradation products, HS released from GPC1, and SYN aggregates (filamentous SYN [SYNfil]). Suppression of HS release from GPC1 by inhibition of ß-secretase or by NO deprivation resulted in no or slight increase in SYNfil aggregation. Stimulation of HS release by ascorbate did not further increase SYNfil staining. Interleukin-6 (IL-6) induced increased APP and GPC1 processing and SYNfil formation, which was reduced when ß-secretase was inhibited and when HS release was impeded by NO deprivation. Ascorbate restored APP and GPC1 processing but did not affect SYNfil formation. Ascorbate-dependent differentiation of NPC resulted in the expression of tyrosine hydroxylase (TH) which colocalized with SYNfil. Suppression of APP processing by inhibition of ß-secretase greatly disturbed the differentiation process. IL-6 induced coclustering of APP-degradation products, TH, HS, and SYNfil, which could be reversed by stimulation of HS release from GPC1 by excess ascorbate. We suggest that continuous release of HS from GPC1 moderates SYN aggregation and supports differentiation of NPC to dopaminergic neurons.


Asunto(s)
Glipicanos , Células-Madre Neurales , Humanos , alfa-Sinucleína , Precursor de Proteína beta-Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Diferenciación Celular , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Interleucina-6 , Células-Madre Neurales/metabolismo
2.
Glycobiology ; 32(4): 333-342, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-34939110

RESUMEN

In Parkinson's disease (PD), there is accumulation of α-synuclein (SYN) aggregates in neurons, which is promoted by neuroinflammation. The cytokines TNF-α, IL-1ß and IL-6 induce accumulation of degradation products of the amyloid precursor protein (APP) combined with heparan sulfate (HS) chains released from glypican-1 (Gpc-1) by NO-dependent cleavage. We have investigated the effects of the cytokines and HS on SYN aggregation and secretion in dividing human neuroblastoma (SH-SY5Y) and inducible neural progenitor cells (NPC) by using immunofluorescence microscopy, vesicle isolation and slot blotting with antibodies recognizing SYN monomers and aggregates, Gpc-1, the released HS, endosomes, and autophagosomes. In SH-SY5Y cells, the capacity to release HS was fully utilized, while NPC displayed dormant capacity. TNF-α induced increased formation of SYN aggregates and clustering of HS in SH-SY5Y cells. When the supply of NO was simultaneously increased, SYN and HS accumulation disappeared. When NO formation was inhibited, SYN and HS aggregation also disappeared, but there was now a 4-fold increase in SYN secretion. In NPC, IL-6 induced increased aggregation of SYN and stimulated HS release from Gpc-1. Both SYN and HS co-localized with autophagosome marker. When HS-deficient Gpc-1 was simultaneously generated, by using a cyanobacterial neurotoxin, accumulation diminished and there was massive secretion of SYN. We suggest that the cytokines increase APP processing, which initiates NO-dependent release of HS from Gpc-1. The APP degradation products also trigger SYN aggregation. As HS can inhibit APP processing, HS- or NO-deficiency may result in autophagosomal dysfunction and both APP degradation products and SYN are secreted.


Asunto(s)
Glipicanos , alfa-Sinucleína , Citocinas/metabolismo , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacología
3.
Glycobiology ; 31(7): 800-811, 2021 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-33403386

RESUMEN

The Apolipoprotein E4 (ApoE4) genotype is the most influential risk factor for sporadic Alzheimer's disease. It appears to be associated with retarded endosome-to-autophagosome trafficking. The amyloid precursor protein (APP) and the heparan sulfate (HS)-containing proteoglycan glypican-1 (Gpc-1) are both processed in endosomes, and mutually regulated by the APP degradation products and the released HS. We have investigated APP and Gpc-1 processing in ApoE3 and ApoE4 expressing human fibroblasts, in human neural stem cells (NSC) exposed to the cholesterol transport inhibitor U18666A and in induced neurons obtained by reprogramming of ApoE fibroblasts (ApoE-iN). We have used immunofluorescence microscopy, flow cytometry, and sodium dodecyl sulfate-polyacrylamide gel electrophoresis western blotting with antibodies recognizing the released HS, APP, amyloid ß(Aß), late endosomes (Rab7), autophagosomes (LC3) and neurons (Tuj1). We found that the capacity to release HS was not fully utilized in ApoE4 expressing fibroblasts and that HS-Aß complexes accumulated in the nuclei. In ApoE3 fibroblasts, the ß-cleaved APP C-terminal fragment (ß-CTF) and Aß were primarily present in late endosomes and autophagosomes. When HS release from Gpc-1 was enhanced by ascorbate in ApoE4/4 fibroblasts, there was efficient transfer of Aß and HS from the nuclei to autophagosomes. In U18666A-treated NSC as well as in ApoE4/4-iN we repeatedly found accumulation of APP degradation products (ß-CTF/Aß). This was reversed by subsequent exposure to ascorbate or dehydroascorbic acid.


Asunto(s)
Enfermedad de Alzheimer , Glipicanos , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Ácido Ascórbico/farmacología , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Humanos
4.
Glycobiology ; 30(8): 539-549, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32039447

RESUMEN

Proinflammatory cytokines stimulate expression of ß-secretase, which increases processing of amyloid precursor protein (APP), ultimately leading to the deposition of amyloid beta (Aß). The N-terminal domain of ß-cleaved APP supports Cu/NO-dependent release of heparan sulfate (HS) from the glypican-1 (Gpc-1) proteoglycan. HS is an inhibitor of ß-secretase, thereby constituting a regulatory, negative feedback loop. Here, we have investigated the effect of the proinflammatory cytokines TNF-α, IL-1ß and IL-6 on the interplay between APP processing and release of HS from Gpc-1 in neuronal cells. We have used deconvolution immunofluorescence microscopy and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and a panel of monoclonal/polyclonal antibodies recognizing the released HS, the N-terminus of Aß, Aß, the C-terminus of APP and the autophagosome marker LC3 as well as the chemical lysosome marker LysoTrackerRed (LTR). We repeatedly found that N2a neuroblastoma cells and human neural stem cells grown in the presence of the cytokines developed large cytoplasmic clusters, which stained positive for HS, the N-terminus of Aß, Aß, the C-terminus of APP, LC3 and LTR, indicating accumulation of HS and APP/APP degradation products in enlarged autophagosomes/lysosomes. The SDS-PAGE of immunoisolates obtained from TNF-α-treated N2a cells by using anti-C-terminus of APP revealed the presence of SDS-stable complexes between HS and the C-terminal fragment of ß-cleaved APP (ßCTF) migrating in the range 10-18 kDa. Clustered accumulation of ßCTF disappeared when HS release was prevented and slightly enhanced when HS release was increased. Hence, when proinflammatory cytokines induce increased processing of APP, inhibition of ß-secretase by HS is insufficient, which may lead to the impaired autophagosomal degradation.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Autofagosomas/metabolismo , Citocinas/metabolismo , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Neuronas/metabolismo , Animales , Línea Celular Tumoral , Ratones
5.
Exp Cell Res ; 379(2): 172-181, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30953622

RESUMEN

The neurotoxin ß-N-methylamino-l-alanine replaces l-serine in proteins and produces Alzheimer-like pathology. In proteoglycans, e.g. glypican-1, this should preclude substitution with heparan sulfate chains. Reduced release of heparan sulfate should increase ß-secretase activity and processing of amyloid precursor protein. Cultured cells were treated with ß-N-methylamino-l-alanine during the growth-phase and the effect on heparan sulfate substitution and amyloid precursor protein processing was evaluated using antibodies specific for heparan sulfate, the N- and C-termini of the C-terminal fragment of ß-cleaved amyloid precursor protein, and amyloid beta followed by immunofluorescence microscopy, flow cytometry or SDS-PAGE. Mouse fibroblasts, N2a neuroblastoma cells and human neural stem cells released less heparan sulfate when grown in the presence of ß-N-methylamino-l-alanine. Cells expressing a recombinant, anchor-less glypican-1 secreted heparan sulfate-deficient glypican-1. There was increased processing of amyloid precursor protein in N2a cells when grown in the presence of the neurotoxin. The degradation products accumulated in cytoplasmic clusters. Secretion of amyloid beta increased approx. 3-fold. Human neural stem cells also developed cytoplasmic clusters containing degradation products of amyloid precursor protein. When non-dividing mouse N2a cells or cortical neurons were exposed to ß-N-methylamino-l-alanine there was no effect on heparan sulfate substitution in glypican-1 or on amyloid precursor protein processing.


Asunto(s)
Aminoácidos Diaminos/farmacología , Precursor de Proteína beta-Amiloide/metabolismo , Endosomas/efectos de los fármacos , Glipicanos/metabolismo , Neuronas/efectos de los fármacos , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/efectos de los fármacos , Animales , Proteínas Portadoras/metabolismo , Toxinas de Cianobacterias , Endosomas/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Proteoglicanos/metabolismo
6.
Exp Cell Res ; 364(2): 133-142, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29408503

RESUMEN

Import of the polyamine spermine from the extracellular environment depends on the presence of cell surface heparan sulfate proteoglycans, such as glypican-1. This proteoglycan is internalized by endocytosis, releases its heparan sulfate chains in endosomes by a nitric oxide-, copper- and amyloid precursor protein-dependent mechanism, then penetrates the membrane and is transported to the nucleus and then to autophagosomes. This process is spontaneous or induced by ascorbate depending on the growth-state of the cell. Here, we have explored possible connections between the heparan sulfate traffic route and spermine uptake and delivery in wild-type and Tg2576 mouse fibroblasts. Cells were examined by deconvolution immunofluorescence microscopy. The antibodies used were specific for spermine, glypican-1-derived heparan sulfate, Rab7, nucleolin and a marker for autophagosomes. Endogenous immunostainable spermine was primarily associated with autophagosomes. When spermine synthesis was inhibited, imported spermine appeared in Rab7-positive endosomes. When ascorbate was added, heparan sulfate and spermine were transported to the nucleus where they colocalized with nucleolin. Spermine also appeared in autophagosomes. In a pulse-chase experiment, heparan sulfate and spermine were first arrested in late endosomes by actinomycin D treatment. During the chase, when arrest was abolished, heparan sulfate and spermine were both transported to the nucleus and targeted nucleolin. In amyloid precursor protein-/--fibroblasts, ascorbate failed to induce release of heparan sulfate and spermine remained in the endosomes. We propose that cell surface glypican-1 carries spermine to the endosomes and that the released heparan sulfate carries spermine across the membrane into the cytosol and then to the nucleus.


Asunto(s)
Endosomas/metabolismo , Fibroblastos/metabolismo , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Espermina/metabolismo , Animales , Fibroblastos/citología , Ratones , Ratones Noqueados
7.
Exp Cell Res ; 360(2): 171-179, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28893506

RESUMEN

Ascorbate-induced release of heparan sulfate from S-nitrosylated heparan sulfate proteoglycan glypican-1 takes place in endosomes. Heparan sulfate penetrates the membrane and is transported to the nucleus. This process is dependent on copper and on expression and processing of the amyloid precursor protein. It remains unclear how exogenously supplied ascorbate can generate HS-anMan in endosomes and how passage through the membrane is facilitated. Here we have examined wild-type, Alzheimer Tg2576 and amyloid precursor protein (-/-) mouse fibroblasts and human fetal and Niemann-Pick C1 fibroblasts by using deconvolution immunofluorescence microscopy, siRNA technology and [S35]sulfate-labeling, vesicle isolation and gel chromatography. We found that ascorbate-induced release of heparan sulfate was dependent on expression of endosomal cytochrome b561. Formation and nuclear transport of heparan sulfate was suppressed by inhibition of ß-processing of the amyloid precursor protein and formation was restored by copper (I) ions. Membrane penetration was not dependent on amyloid beta channel formation. Inhibition of endosomal exit resulted in accumulation of heparan sulfate in vesicles that exposed the C-terminal of the amyloid precursor protein externally. Endosome-to-nucleus transport was also dependent on expression of the Niemann-Pick C1 protein. We propose that ascorbate is taken up from the medium and is oxidized by cytochrome b561 which, in turn, reduces copper (II) to copper (I) present in the N-terminal, ß-cleaved domain of the amyloid precursor protein. Re-oxidation of copper (I) is coupled to reductive, deaminative release of heparan sulfate from glypican-1. Passage through the membrane may be facilitated by the C-terminal, ß-cleaved fragment of the amyloid precursor protein and the Niemann-Pick C1 protein.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Ácido Ascórbico/farmacología , Proteínas Portadoras/fisiología , Cobre/fisiología , Grupo Citocromo b/fisiología , Endosomas/metabolismo , Glipicanos/metabolismo , Glicoproteínas de Membrana/fisiología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Células Cultivadas , Endosomas/efectos de los fármacos , Heparitina Sulfato , Humanos , Péptidos y Proteínas de Señalización Intracelular , Membranas/efectos de los fármacos , Membranas/metabolismo , Ratones , Ratones Transgénicos , Proteína Niemann-Pick C1 , Nitrosación , Procesamiento Proteico-Postraduccional
8.
Exp Cell Res ; 354(1): 31-39, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28300561

RESUMEN

The recycling, S-nitrosylated heparan sulfate (HS) proteoglycan glypican-1 releases anhydromannose (anMan)-containing HS chains by a nitrosothiol-catalyzed cleavage in endosomes that can be constitutive or induced by ascorbate. The HS-anMan chains are then transported to the nucleus. A specific nuclear target for HS-anMan has not been identified. We have monitored endosome-to-nucleus trafficking of HS-anMan by deconvolution and confocal immunofluorescence microscopy using an anMan-specific monoclonal antibody in non-growing, ascorbate-treated, and growing, untreated, wild-type mouse embryonic fibroblasts and hypoxia-exposed Alzheimer mouse Tg2576 fibroblasts and human U87 glioblastoma cells. In all cells, nuclear HS-anMan targeted a limited number of sites of variable size where it colocalized with DNA and nucleolin, an established marker for nucleoli. HS-anMan also colocalized with ethynyl uridine-tagged nascent RNA and two acetylated forms of histone H3. Acute hypoxia increased the formation of HS-anMan in both Tg2576 and U87 cells. A portion of HS-anMan colocalized with nucleolin at small discrete sites, while most of the nucleolin and nascent RNA was dispersed. In U87 cells, HS-anMan, nucleolin and nascent RNA reassembled after prolonged hypoxia. Nucleolar HS may modulate synthesis and/or release of rRNA.


Asunto(s)
Enfermedad de Alzheimer/genética , Núcleo Celular/metabolismo , Glipicanos/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Núcleo Celular/ultraestructura , Modelos Animales de Enfermedad , Endosomas/metabolismo , Endosomas/ultraestructura , Heparitina Sulfato/metabolismo , Humanos , Manosa/genética , Manosa/metabolismo , Ratones , Microscopía Confocal , Óxido Nítrico/metabolismo , Fosfoproteínas/ultraestructura , Proteínas de Unión al ARN/ultraestructura , Nucleolina
9.
Glycobiology ; 26(6): 623-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26791445

RESUMEN

There is a functional relationship between the heparan sulfate proteoglycan glypican-1 and the amyloid precursor protein (APP) of Alzheimer disease. In wild-type mouse embryonic fibroblasts, expression and processing of the APP is required for endosome-to-nucleus translocation of anhydromannose-containing heparan sulfate released from S-nitrosylated glypican-1 by ascorbate-induced, nitrosothiol-catalyzed deaminative cleavage. In fibroblasts from the transgenic Alzheimer mouse Tg2576, there is increased processing of the APP to amyloid-ß peptides. Simultaneously, there is spontaneous formation of anhydromannose-containing heparan sulfate by an unknown mechanism. We have explored the effect of hypoxia on anhydromannose-containing heparan sulfate formation in wild-type and Tg2576 fibroblasts by deconvolution immunofluorescence microscopy and flow cytometry using an anhydromannose-specific monoclonal antibody and by (35)SO4-labeling experiments. Hypoxia prevented ascorbate-induced heparan sulfate release in wild-type fibroblasts, but induced an increased formation of anhydromannose-positive and (35)S-labeled heparan sulfate in Tg2576 fibroblasts. This appeared to be independent of glypican-1 S-nitrosylation as demonstrated by using a monoclonal antibody specific for S-nitrosylated glypican-1. In hypoxic wild-type fibroblasts, addition of nitrite to the medium restored anhydromannose-containing heparan sulfate formation. The increased release of anhydromannose-containing heparan sulfate in hypoxic Tg2576 fibroblasts did not require addition of nitrite. However, it was suppressed by inhibition of the nitrite reductase activity of xanthine oxidoreductase/aldehyde oxidase or by inhibition of p38 mitogen-activated protein kinase or by chelation of iron. We propose that normoxic Tg2576 fibroblasts maintain a high level of anhydromannose-containing heparan sulfate production by a stress-activated generation of nitric oxide from endogenous nitrite. This activation is enhanced by hypoxia.


Asunto(s)
Fibroblastos/efectos de los fármacos , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Manosa/análogos & derivados , Óxido Nítrico/farmacología , Oxígeno/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Anticuerpos Monoclonales/química , Ácido Ascórbico/farmacología , Hipoxia de la Célula , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Glipicanos/química , Heparitina Sulfato/química , Humanos , Quelantes del Hierro/farmacología , Manosa/química , Manosa/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Óxido Nítrico/metabolismo , Nitrito Reductasas/antagonistas & inhibidores , Nitrito Reductasas/metabolismo , Nitritos/química , Nitritos/farmacología , Oxidación-Reducción , Cultivo Primario de Células , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
J Biol Chem ; 289(30): 20871-8, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24898256

RESUMEN

Anhydromannose (anMan)-containing heparan sulfate (HS) derived from the proteoglycan glypican-1 is generated in endosomes by an endogenously or ascorbate-induced S-nitrosothiolcatalyzed reaction. Processing of the amyloid precursor protein (APP) and APP-like protein 2 (APLP2) by ß- and γ-secretases into amyloid ß(A) and Aß-like peptides also takes place in these compartments. Moreover, anMan-containing HS suppresses the formation of toxic Aß assemblies in vitro. We showed by using deconvolution immunofluorescence microscopy with an anMan-specific monoclonal antibody as well as (35)S labeling experiments that expression of APP/APLP2 is required for ascorbate-induced transport of HS from endosomes to the nucleus. Nuclear translocation was observed in wild-type mouse embryonic fibroblasts (WT MEFs), Tg2576 MEFs, and N2a neuroblastoma cells but not in APP(-/-) and APLP2(-/-) MEFs. Transfection of APP(-/-) cells with a vector encoding APP restored nuclear import of anMan-containing HS. In WT MEFs and N2a neuroblastoma cells exposed to ß- or γ-secretase inhibitors, nuclear translocation was greatly impeded, suggesting involvement of APP/APLP2 degradation products. In Tg2576 MEFs, the ß-inhibitor blocked transport, but the γ-inhibitor did not. During chase in ascorbate- free medium, anMan-containing HS disappeared from the nuclei of WT MEFs. Confocal immunofluorescence microscopy showed that they appeared in acidic, LC3-positive vesicles in keeping with an autophagosomal location. There was increased accumulation of anMan-containing HS in nuclei and cytosolic vesicles upon treatment with chloroquine, indicating that HS was degraded in lysosomes. Manipulations of APP expression and processing may have deleterious effects upon HS function in the nucleus.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Núcleo Celular/metabolismo , Endosomas/metabolismo , Regulación de la Expresión Génica/fisiología , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular Tumoral , Núcleo Celular/genética , Endosomas/genética , Glipicanos/genética , Ratones , Ratones Noqueados , Fagosomas/genética , Fagosomas/metabolismo
11.
Glycobiology ; 25(5): 548-56, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25527428

RESUMEN

Anhydromannose (anMan)-containing heparan sulfate (HS) derived from S-nitrosylated glypican-1 is generated in endosomes by an endogenously or ascorbate induced S-nitrosothiol-catalyzed reaction. Expression and processing of amyloid precursor protein (APP) is required to initiate formation and endosome-to-nucleus translocation of anMan-containing HS in wild-type mouse embryonic fibroblasts (WT MEF). HS is then transported to autophagosomes and finally degraded in lysosomes. To investigate how APP-derived amyloid ß (Aß) peptide affects intracellular trafficking of HS, we have studied nuclear transit as well as autophagosome/lysosome targeting and degradation in transgenic Alzheimer disease mouse (Tg2576) MEF which produce increased amounts of Aß. Deconvolution immunofluorescence microscopy with an anMan-specific monoclonal antibody showed anMan staining in the nuclei of Tg2576 MEF after 5 min of ascorbate treatment and after 15 min in WT MEF. There was also greater nuclear accumulation of HS in Tg2576 MEF as determined by (35)S-sulfate-labeling experiments. Tg2576 MEF was less sensitive to inhibition of NO production and copper-chelation than WT MEF. By using APP- and Aß-recognizing antibodies, we observed nuclear translocation of Aß peptide in Tg2576 MEF but not in WT MEF. HS remained in the nucleus of WT MEF for at least 8 h and was then transported to autophagosomes. By 8 h, HS had disappeared from the nuclei of Tg2576 MEF but colocalized poorly with the autophagosome marker LC3. Aß also disappeared rapidly from the nuclei of Tg2576 MEF. Initially, it appeared in acidic vesicles and later it accumulated extracellularly. Thus, in Tg2576 MEF there is nuclear accumulation as well as secretion of Aß and impaired degradation of HS.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Núcleo Celular/metabolismo , Fibroblastos/metabolismo , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Lisosomas/metabolismo , Ratones , Proteolisis
12.
Glycoconj J ; 32(9): 675-84, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26318599

RESUMEN

Heparan sulfate (HS)-containing, S-nitrosylated (SNO) glypican-1 (Gpc-1) releases anhydromannose-containing HS (anMan-HS) by SNO-catalyzed autodegradation in endosomes. Transport of anMan-HS to the nucleus requires processing of the amyloid precursor protein (APP) to amyloid beta peptides (Aß). To further examine the relationship between APP and Gpc-1 processing in normal fibroblasts we have suppressed Gpc-1 autodegradation by aminoguanidine inhibition of NO synthesis and prevented lysosomal degradation of anMan-HS by using chloroquine. Deconvolution immunofluorescence microscopy and SDS-PAGE using anMan- and APP/Aß-specific antibodies and markers for nuclei and autophagosomes were used to identify subcellular localization of Aß and its oligomeric state. Wild-type mouse embryonic fibroblasts (WT MEF) grown during NO-deprivation accumulated 95-98% of Aß as oligomers in the nucleus. WT MEF treated with chloroquine accumulated both anMan-HS and Aß, first in the nucleus then in autophagosomes. Maximal nuclear anMan-HS and Aß accumulation was obtained after 4 and 7 h of growth, respectively. Both yielded similar banding patterns on SDS-PAGE which were also similar to the Aß oligomers obtained after NO-deprivation. Nuclear Aß accumulation was marginally increased (from 54 to 58%) by suppression of both release and degradation of anMan-HS. Nuclear exit of Aß, accumulated during growth in aminoguanidine, was enhanced by ascorbate-induced reactivation of anMan-HS production. Transgenic Alzheimer disease mouse (Tg2576) MEF, which produces excess amount of Aß was used for comparison. Overall, nuclear Aß exit and lysosomal degradation was compromised by inhibition of the autophagosome-lysosome pathway in both WT and Tg2576 MEF, while only WT MEF was sensitive to suppression of Gpc-1 autodegradation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Fibroblastos/metabolismo , Glipicanos/metabolismo , Óxido Nítrico/deficiencia , Proteolisis , Animales , Línea Celular , Núcleo Celular/metabolismo , Ratones , Óxido Nítrico/metabolismo
13.
Neuroscience ; 553: 121-127, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38992568

RESUMEN

INTRODUCTION: Alzheimer's disease (AD) is characterized by accumulation of amyloid beta (Aß) and hyperphosphorylated tau (Tau-P) in the brain. Aß enhances the activity of kinases involved in the formation of Tau-P. Phosphorylation at Thr 181 determines the propagation of multiple tau phosphorylations. Aß is derived from the amyloid precursor protein (APP). Cleavage of APP by ß-secretase also initiates release of heparan sulfate (HS) from the proteoglycan glypican-1 (GPC1). OBJECTIVES: In this study, we have explored possible connections between GPC1 expression, HS release, APP processing and Tau-P formation in human neural stem cells. METHODS: GPC1 formation was suppressed by using CRISPR/Cas9 and increased by using a vector encoding GPC1. HS release from GPC1 was increased by growing cells in medium containing Arg and ascorbate. Effects were monitored by immunofluorescence microscopy and slot immunoblotting using antibodies/antisera recognizing Aß, GPC1, HS released from GPC1, total Tau, and Tau phosphorylated at Thr-181, 217 or 231. The latter have been used as blood biomarkers for AD. RESULTS: Suppression of GPC1 expression resulted in increased phosphorylation at Thr 181 and Thr 217. When GPC1 was overexpressed, phosphorylation at Thr 217 decreased. Stimulation of HS release from GPC1 diminished tau phosphorylation at all of the three Thr positions, while expression of GPC1 was unaffected. Simultaneous stimulation of HS release and APP processing by the cytokine TNF-α also suppressed tau phosphorylation. CONCLUSION: The increased release of GPC1-derived HS may interfere with Aß formation and/or Aß interaction with tau.


Asunto(s)
Péptidos beta-Amiloides , Glipicanos , Células-Madre Neurales , Proteínas tau , Humanos , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Glipicanos/metabolismo , Células-Madre Neurales/metabolismo , Fosforilación/fisiología , Proteínas tau/metabolismo
14.
Glycobiology ; 23(12): 1510-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24026238

RESUMEN

The amyloid beta (Aß) peptides (mainly Aß40 and Aß42), which are derived from the amyloid precursor protein (APP), can oligomerize into antibody A11-positive, neurotoxic species, believed to be involved in Alzheimer's disease. Interestingly, APP binds strongly to the heparan sulfate (HS) proteoglycan (PG) glypican-1 (Gpc-1) in vitro and both proteins are colocalized inside cells. In endosomes, APP is proteolytically processed to yield Aß peptides. The HS chains of S-nitrosylated (SNO) Gpc-1 PG are cleaved into anhydromannose (anMan)-containing di- and oligosaccharides by an NO-dependent reaction in the same compartments. Here, we have studied the toxicity of oligomers/aggregates of Aß40 and Aß42, as well as Aß40/42 mixtures that were formed in the presence of immobilized Gpc-1 PG or immobilized HS oligosaccharides. Afterwards, Aß was displaced from the matrices, analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis and assayed for A11 immunoreactivity, for effects on growth of mouse N2a neuroblastoma cells and for membrane leakage in rat cortical neurons. HS generally promoted and accelerated Aß multimerization into oligomers as well as larger aggregates that were mostly A11 positive and showed toxic effects. However, non-toxic Aß was formed in the presence of Gpc-1 PG or when anMan-containing HS degradation products were simultaneously generated. Both toxic and non-toxic Aß peptides were taken up by the cells but toxic forms appeared to enter the nuclei to a larger extent. The protection afforded by the presence of HS degradation products may reflect a normal intracellular function for the Aß peptides.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Péptidos beta-Amiloides/metabolismo , Glipicanos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Péptidos beta-Amiloides/farmacología , Péptidos beta-Amiloides/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/citología , Desaminación , Humanos , Ratones , Neuroblastoma/patología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar
15.
J Biol Chem ; 286(31): 27559-72, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21642435

RESUMEN

Amyloid ß (Aß) is generated from the copper- and heparan sulfate (HS)-binding amyloid precursor protein (APP) by proteolytic processing. APP supports S-nitrosylation of the HS proteoglycan glypican-1 (Gpc-1). In the presence of ascorbate, there is NO-catalyzed release of anhydromannose (anMan)-containing oligosaccharides from Gpc-1-nitrosothiol. We investigated whether these oligosaccharides interact with Aß during APP processing and plaque formation. anMan immunoreactivity was detected in amyloid plaques of Alzheimer (AD) and APP transgenic (Tg2576) mouse brains by immunofluorescence microscopy. APP/APP degradation products detected by antibodies to the C terminus of APP, but not Aß oligomers detected by the anti-Aß A11 antibody, colocalized with anMan immunoreactivity in Tg2576 fibroblasts. A 50-55-kDa anionic, sodium dodecyl sulfate-stable, anMan- and Aß-immunoreactive species was obtained from Tg2576 fibroblasts using immunoprecipitation with anti-APP (C terminus). anMan-containing HS oligo- and disaccharide preparations modulated or suppressed A11 immunoreactivity and oligomerization of Aß42 peptide in an in vitro assay. A11 immunoreactivity increased in Tg2576 fibroblasts when Gpc-1 autoprocessing was inhibited by 3-ß[2(diethylamino)ethoxy]androst-5-en-17-one (U18666A) and decreased when Gpc-1 autoprocessing was stimulated by ascorbate. Neither overexpression of Gpc-1 in Tg2576 fibroblasts nor addition of copper ion and NO donor to hippocampal slices from 3xTg-AD mice affected A11 immunoreactivity levels. However, A11 immunoreactivity was greatly suppressed by the subsequent addition of ascorbate. We speculate that temporary interaction between the Aß domain and small, anMan-containing oligosaccharides may preclude formation of toxic Aß oligomers. A portion of the oligosaccharides are co-secreted with the Aß peptides and deposited in plaques. These results support the notion that an inadequate supply of vitamin C could contribute to late onset AD in humans.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Ácido Ascórbico/farmacología , Glipicanos/química , Heparitina Sulfato/inmunología , Óxido Nítrico/química , Animales , Catálisis , Citometría de Flujo , Humanos , Hidrólisis , Ratones , Ratones Transgénicos , Microscopía Fluorescente
16.
Glycobiology ; 22(11): 1480-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22801553

RESUMEN

The membrane lipid-anchored glypicans (Gpcs) [heparan sulfate (HS) proteoglycans (PGs)] are present in both vertebrates and invertebrates and serve as important modulators of growth factors and morphogens during development. Their core proteins are similar and consist of a large N-terminal domain comprising 14 evolutionary conserved cysteines and a C-terminal stalk carrying the HS side chains and the lipid anchor. Cysteines in Gpc-1 can be S-nitrosylated but their positions have not been identified. The recently determined crystal structure of the N-terminal domain of Gpc-1 has revealed that all the evolutionary conserved cysteines form intramolecular disulfide bonds. However, Gpc-1 contains two more, non-conserved cysteines in the C-terminal stalk, located near the HS attachment sites. We show here that the non-conserved cysteines are free thiols as a Gpc-1 core protein containing the C-terminal stalk could be biotinylated by 1-biotinamido-4-(4'-[maleimidomethyl-cyclohexane]-carboxyamido)butane. After S-nitrosylation by using a nitric oxide (NO) donor and copper ions, the Gpc-1 core protein was retained on an affinity matrix substituted with HS oligosaccharides containing N-unsubstituted glucosamines (GlcNH(2)/NH(3)(+)). The protein was displaced with 0.2 M glucosamine but also by 2 mM ascorbate. In the latter case, the HS of the affinity matrix was simultaneously cleaved into fragments containing anhydromannose (anMan). We propose that the S-nitrosocysteine residues interact with closely located GlcNH(2)/NH(3)(+) in the HS side chains of the Gpc-1 PG. Addition of ascorbate induces a series of reactions that eventually releases HS fragments with reducing terminal anMan, presumably without the formation of free NO.


Asunto(s)
Cisteína/análogos & derivados , Glucosamina/química , Glipicanos/química , Heparitina Sulfato/química , S-Nitrosotioles/química , Cisteína/química , Humanos
17.
Eur J Neurosci ; 28(5): 964-72, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18717736

RESUMEN

The copper-binding cellular prion protein (PrP(C)) and the heparan sulphate (HS)-containing proteoglycan glypican-1 (Gpc-1) can both be attached to lipid rafts via their glycosylphosphatidylinositol anchors, and copper ions stimulate their cointernalization from the cell surface to endosomes. The prion protein controls cointernalization and delivers copper necessary for S-nitrosylation of conserved cysteines in the Gpc-1 core protein. Later, during recycling through endosomal compartments, nitric oxide can be released from the S-nitroso groups and catalyses deaminative degradation and release of the HS substituents. Here, by using confocal immunofluorescence microscopy, we show that normal PrP(C) and Gpc-1 colocalize inside GT1-1 cells. However, in scrapie-infected cells (ScGT1-1), Gpc-1 protein remained at the cell surface separate from the cellular prion protein. Scrapie infection stimulated Gpc-1 autoprocessing and the generated HS degradation products colocalized with intracellular aggregates of the disease-related scrapie prion protein isoform (PrP(Sc)). Coimmunoprecipitation experiments demonstrated an association between Gpc-1 and PrP(C) in uninfected cells, and between HS degradation products and PrP(Sc) in infected cells. Silencing of Gpc-1 expression or prevention of Gpc-1 autoprocessing elevated the levels of intracellular PrP(Sc) aggregates in infected cells. These results suggest a role for Gpc-1 autoprocessing in the clearance of PrP(Sc) from infected cells.


Asunto(s)
Encéfalo/metabolismo , Glipicanos/metabolismo , Neuronas/metabolismo , Priones/metabolismo , Scrapie/metabolismo , Animales , Encéfalo/fisiopatología , Compartimento Celular/fisiología , Extractos Celulares , Membrana Celular/metabolismo , Células Cultivadas , Endocitosis/fisiología , Cuerpos de Inclusión/metabolismo , Ratones , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Transporte de Proteínas/fisiología , Scrapie/fisiopatología
18.
Trends Mol Med ; 13(4): 143-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17344097

RESUMEN

The reduced form of vitamin C, ascorbic acid, is well known for its function as an antioxidant and as a protective agent against scurvy. However, many recent studies indicate other functions for vitamin C in mammalian cells. Novel findings provide possible explanations for observed beneficial effects of a high intake of vitamin C on cell growth, gene transcription, host resistance to infection, uptake of polyamines and clearance of misfolded proteins. Vitamin C exerts its effects indirectly via hypoxia-inducible factor, nitric oxide synthase and the heparan sulfate proteoglycan glypican-1, which is deglycanated in a vitamin C- and copper-dependent reaction.


Asunto(s)
Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacología , Glipicanos/metabolismo , Animales , Antioxidantes/administración & dosificación , Antioxidantes/metabolismo , Antioxidantes/farmacología , Ácido Ascórbico/administración & dosificación , Humanos , Modelos Biológicos , Óxido Nítrico/metabolismo , Necesidades Nutricionales , Oxígeno/metabolismo
19.
Int J Biochem Cell Biol ; 35(2): 125-9, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12479862

RESUMEN

A family of lipid-linked heparan sulfate (HS) proteoglycans, later named glypicans, were identified some 15 years ago. The discoveries that mutations in genes involved in glypican assembly cause developmental defects have brought them into focus. Glypicans have a characteristic pattern of 14 conserved cysteine residues. There are also two-three attachment sites for HS side-chains near the membrane anchor. The HS side-chains consist of a repeating disaccharide back-bone that is regionally and variably modified by epimerization and different types of sulfations, creating a variety of binding sites for polycationic molecules, especially growth factors. Recycling forms of glypican-1 are potential vehicles for transport of cargo into and through cells. The glypican-1 core protein is S-nitrosylated and nitric oxide released from these sites cleave the HS chains at glucosamine units lacking N-substitution. This processing is necessary for polyamine uptake.


Asunto(s)
Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Animales , Transporte Biológico , Regulación del Desarrollo de la Expresión Génica , Sustancias de Crecimiento/metabolismo , Proteoglicanos de Heparán Sulfato/química , Heparitina Sulfato/metabolismo , Humanos , Óxido Nítrico/metabolismo , Poliaminas/metabolismo
20.
Cancer Res ; 70(9): 3771-9, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20406966

RESUMEN

Glycosaminoglycan (GAG) chains anchored to core proteins form proteoglycans, widely distributed cell-surface macromolecules with multiple functions, such as regulation of growth factor and cytokine signaling, cell-cell interactions, and uptake of biomolecules. The biosynthesis of GAG can be manipulated by xylosides attached to various hydrophobic groups, and we have earlier reported that a naphthoxyloside, 2-(6-hydroxynaphthyl) beta-D-xylopyranoside (XylNapOH), which serves as a primer for GAG synthesis, reduces tumor load up to 97% in vivo, despite lower efficiency in vitro. Here we show, using radiolabeled xylosides and coculture experiments, that XylNapOH-treated bladder and breast carcinoma cells secrete antiproliferative GAG chains that are taken up by both normal and cancer cells and transported to the cell nuclei where they induce an antiproliferative effect, accompanied by apoptosis. We also show that XylNapOH treatment lowers the level of histone H3 acetylation selectively in bladder and breast carcinoma cells without affecting expression of histone H3. However, XylNapOH-primed GAG chains from normal cells are not internalized and do not cause growth retardation. Using in vitro and in vivo C6 glioma cell and tumor models, we show that XylNapOH is much more effective in vivo than in vitro. We propose that, in vivo, the antiproliferative XylNapOH-primed GAG chains produced by tumor cells inhibit tumor growth in an autocrine fashion by formation of antiproliferative GAG chains on the xyloside prodrug, whereas no antiproliferative GAG chains are produced by surrounding normal cells. This is a novel mechanism for targeting tumor cells, making these xylosides promising drug candidates for antitumor therapy.


Asunto(s)
Glicosaminoglicanos/metabolismo , Histonas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Acetilación/efectos de los fármacos , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Femenino , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/patología , Glicosaminoglicanos/biosíntesis , Glicosaminoglicanos/farmacocinética , Glicosaminoglicanos/farmacología , Glicósidos/metabolismo , Glicósidos/farmacocinética , Glicósidos/farmacología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/tratamiento farmacológico , Ratas , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA