Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(7): e0159119, 2016.
Article in English | MEDLINE | ID: mdl-27447728

ABSTRACT

The cellular prion protein (PrPC) has been proposed to play an important role in the pathogenesis of Alzheimer's disease. In cellular models PrPC inhibited the action of the ß-secretase BACE1 on wild type amyloid precursor protein resulting in a reduction in amyloid-ß (Aß) peptides. Here we have assessed the effect of genetic ablation of PrPC in transgenic mice expressing human wild type amyloid precursor protein (line I5). Deletion of PrPC had no effect on the α- and ß-secretase proteolysis of the amyloid precursor protein (APP) nor on the amount of Aß38, Aß40 or Aß42 in the brains of the mice. In addition, ablation of PrPC did not alter Aß deposition or histopathology phenotype in this transgenic model. Thus using this transgenic model we could not provide evidence to support the hypothesis that PrPC regulates Aß production.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Prion Proteins/physiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/pathology , Female , Gene Deletion , Humans , Mice , Mice, Transgenic , Phenotype , Plaque, Amyloid/pathology , Prion Proteins/genetics , Proteolysis
2.
PLoS One ; 8(10): e78806, 2013.
Article in English | MEDLINE | ID: mdl-24205320

ABSTRACT

Bridging integrator 1 (BIN1) has been implicated in sporadic Alzheimer's disease (AD) by a number of genome wide association studies (GWAS) in a variety of populations. Here we measured BIN1 in frontal cortex samples from 24 sporadic AD and 24 age-matched non-dementia brains and correlated the expression of this protein with markers of AD. BIN1 was reduced by 87% (p=0.007) in sporadic AD compared to non-dementia controls, but BIN1 in sporadic AD did not correlate with soluble Aß (r(s)=-0.084, p=0.698), insoluble Aß (r(s)=0.237, p=0.269), Aß plaque load (r(s)=0.063, p=0.771) or phospho-tau load (r(s)=-0.160, p=0.489). In contrast to our findings in sporadic AD, BIN1 was unchanged in the hippocampus from 6 cases of familial AD compared to 6 age-matched controls (p=0.488). BIN1 declined with age in a cohort of non-dementia control cases between 25 and 88 years but the correlation was not significant (rs=-0.449, p=0.081). Although BIN1 is known to have a role in endocytosis, and the processing of the amyloid precursor protein (APP) to form amyloid-ß (Aß) peptides is dependent on endocytosis, knockdown of BIN1 by targeted siRNA or the overexpression of BIN1 in a human neuroblastoma cell line (SH-SY5Y) had no effect on APP processing. These data suggest that the alteration in BIN1 is involved in the pathogenesis of sporadic, but not familial AD and is not a consequence of AD neurodegeneration or the ageing process, a finding in keeping with the numerous GWAS that implicate BIN1 in sporadic AD. However, the mechanism of its contribution remains to be established.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging/metabolism , Alzheimer Disease/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Cell Line, Tumor , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Male , Middle Aged , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
3.
PLoS One ; 8(4): e59554, 2013.
Article in English | MEDLINE | ID: mdl-23577068

ABSTRACT

The cellular prion protein (PrP(C)) has been implicated in the development of Alzheimer's disease (AD). PrP(C) decreases amyloid-ß (Aß) production, which is involved in AD pathogenesis, by inhibiting ß-secretase (BACE1) activity. Contactin 5 (CNTN5) has also been implicated in the development of AD by a genome-wide association study. Here we measured PrP(C) and CNTN5 in frontal cortex samples from 24 sporadic AD and 24 age-matched control brains and correlated the expression of these proteins with markers of AD. PrP(C) was decreased in sporadic AD compared to controls (by 49%, p = 0.014) but there was no difference in CNTN5 between sporadic AD and controls (p = 0.217). PrP(C) significantly inversely correlated with BACE1 activity (rs = -0.358, p = 0.006), Aß load (rs = -0.456, p = 0.001), soluble Aß (rs = -0.283, p = 0.026) and insoluble Aß (rs = -0.353, p = 0.007) and PrP(C) also significantly inversely correlated with the stage of disease, as indicated by Braak tangle stage (rs = -0.377, p = 0.007). CNTN5 did not correlate with Aß load (rs = 0.040, p = 0.393), soluble Aß (rs = 0.113, p = 0.223) or insoluble Aß (rs = 0.169, p = 0.125). PrP(C) was also measured in frontal cortex samples from 9 Down's syndrome (DS) and 8 age-matched control brains. In contrast to sporadic AD, there was no difference in PrP(C) in the DS brains compared to controls (p = 0.625). These data are consistent with a role for PrP(C) in regulating Aß production and indicate that brain PrP(C) level may be important in influencing the onset and progression of sporadic AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Prions/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Brain/enzymology , Brain/pathology , Case-Control Studies , Contactins/metabolism , Down Syndrome/metabolism , Female , Frontal Lobe/metabolism , Humans , Male , Middle Aged
4.
Nat Commun ; 3: 1134, 2012.
Article in English | MEDLINE | ID: mdl-23072804

ABSTRACT

Zinc is released into the synaptic cleft upon exocytotic stimuli, although the mechanism for its reuptake into neurons is unresolved. Here we show that the cellular prion protein enhances the uptake of zinc into neuronal cells. This prion-protein-mediated zinc influx requires the octapeptide repeats and amino-terminal polybasic region in the prion protein, but not its endocytosis. Selective antagonists of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptors block the prion protein-mediated zinc uptake, and the prion protein co-immunoprecipitates with both GluA1 and GluA2 AMPA receptor subunits. Zinc-sensitive intracellular tyrosine phosphatase activity is decreased in cells expressing prion protein and increased in the brains of prion-protein-null mice, providing evidence of a physiological consequence of this process. Prion protein-mediated zinc uptake is ablated in cells expressing familial associated mutants of the protein and in prion-infected cells. These data suggest that alterations in the cellular prion protein-mediated zinc uptake may contribute to neurodegeneration in prion and other neurodegenerative diseases.


Subject(s)
Endocytosis , Neurons/metabolism , Prions/metabolism , Zinc/metabolism , Animals , Cell Line, Tumor , GPI-Linked Proteins/metabolism , Humans , Mice , Nerve Tissue Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Prion Proteins , Prions/chemistry , Protein Subunits/metabolism , Protein Tyrosine Phosphatases/metabolism , Rats , Receptors, AMPA/metabolism , Transfection
5.
Prion ; 6(3): 217-22, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22449984

ABSTRACT

Alzheimer disease (AD) is characterized by the amyloidogenic processing of the amyloid precursor protein (APP), culminating in the accumulation of amyloid-ß peptides in the brain. The enzymatic action of the ß-secretase, BACE1 is the rate-limiting step in this amyloidogenic processing of APP. BACE1 cleavage of wild-type APP (APPWT) is inhibited by the cellular prion protein (PrP (C) ). Our recent study has revealed the molecular and cellular mechanisms behind this observation by showing that PrP (C) directly interacts with the pro-domain of BACE1 in the trans-Golgi network (TGN), decreasing the amount of BACE1 at the cell surface and in endosomes where it cleaves APPWT, while increasing BACE1 in the TGN where it preferentially cleaves APP with the Swedish mutation (APPSwe). PrP (C) deletion in transgenic mice expressing the Swedish and Indiana familial mutations (APPSwe,Ind) failed to affect amyloid-ß accumulation, which is explained by the differential subcellular sites of action of BACE1 toward APPWT and APPSwe. This, together with our observation that PrP (C) is reduced in sporadic but not familial AD brain, suggests that PrP (C) plays a key protective role against sporadic AD. It also highlights the need for an APPWT transgenic mouse model to understand the molecular and cellular mechanisms underlying sporadic AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , PrPC Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Brain/metabolism , Humans , Mice
6.
PLoS One ; 7(2): e31754, 2012.
Article in English | MEDLINE | ID: mdl-22363722

ABSTRACT

There is increasing evidence of molecular and cellular links between Alzheimer's disease (AD) and prion diseases. The cellular prion protein, PrP(C), modulates the post-translational processing of the AD amyloid precursor protein (APP), through its inhibition of the ß-secretase BACE1, and oligomers of amyloid-ß bind to PrP(C) which may mediate amyloid-ß neurotoxicity. In addition, the APP intracellular domain (AICD), which acts as a transcriptional regulator, has been reported to control the expression of PrP(C). Through the use of transgenic mice, cell culture models and manipulation of APP expression and processing, this study aimed to clarify the role of AICD in regulating PrP(C). Over-expression of the three major isoforms of human APP (APP(695), APP(751) and APP(770)) in cultured neuronal and non-neuronal cells had no effect on the level of endogenous PrP(C). Furthermore, analysis of brain tissue from transgenic mice over-expressing either wild type or familial AD associated mutant human APP revealed unaltered PrP(C) levels. Knockdown of endogenous APP expression in cells by siRNA or inhibition of γ-secretase activity also had no effect on PrP(C) levels. Overall, we did not detect any significant difference in the expression of PrP(C) in any of the cell or animal-based paradigms considered, indicating that the control of cellular PrP(C) levels by AICD is not as straightforward as previously suggested.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Prions/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Gene Silencing , Humans , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
7.
J Biol Chem ; 286(38): 33489-500, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21795680

ABSTRACT

In Alzheimer disease amyloid-ß (Aß) peptides derived from the amyloid precursor protein (APP) accumulate in the brain. Cleavage of APP by the ß-secretase BACE1 is the rate-limiting step in the production of Aß. We have reported previously that the cellular prion protein (PrP(C)) inhibited the action of BACE1 toward human wild type APP (APP(WT)) in cellular models and that the levels of endogenous murine Aß were significantly increased in PrP(C)-null mouse brain. Here we investigated the molecular and cellular mechanisms underlying this observation. PrP(C) interacted directly with the prodomain of the immature Golgi-localized form of BACE1. This interaction decreased BACE1 at the cell surface and in endosomes where it preferentially cleaves APP(WT) but increased it in the Golgi where it preferentially cleaves APP with the Swedish mutation (APP(Swe)). In transgenic mice expressing human APP with the Swedish and Indiana familial mutations (APP(Swe,Ind)), PrP(C) deletion had no influence on APP proteolytic processing, Aß plaque deposition, or levels of soluble Aß or Aß oligomers. In cells, although PrP(C) inhibited the action of BACE1 on APP(WT), it did not inhibit BACE1 activity toward APP(Swe). The differential subcellular location of the BACE1 cleavage of APP(Swe) relative to APP(WT) provides an explanation for the failure of PrP(C) deletion to affect Aß accumulation in APP(Swe,Ind) mice. Thus, although PrP(C) exerts no control on cleavage of APP(Swe) by BACE1, it has a profound influence on the cleavage of APP(WT), suggesting that PrP(C) may be a key protective player against sporadic Alzheimer disease.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Prions/metabolism , Amino Acid Sequence , Amyloid Precursor Protein Secretases/chemistry , Animals , Aspartic Acid Endopeptidases/chemistry , Binding Sites , Cell Line , Gene Deletion , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/metabolism , Subcellular Fractions/metabolism
8.
J Alzheimers Dis ; 22(3): 1023-31, 2010.
Article in English | MEDLINE | ID: mdl-20930299

ABSTRACT

The cellular form of the prion protein (PrPC) has been shown to inhibit the production of amyloid-ß which is critically involved in the pathogenesis of Alzheimer's disease (AD). We examined the expression of PrPC by immunoblot analysis in the hippocampus and temporal cortex in sporadic AD, familial AD, and appropriate age-matched controls, and in an aging series (age 20 to 88 years) of brains. PrPC was reduced by 53% (p=0.032) in the hippocampus in sporadic AD as compared to the age-matched controls. No such reduction in PrPC was seen in familial AD. PrPC was reduced in the hippocampus with aging (rs=0.03). The reduction in PrPC in sporadic but not familial AD suggests that reduced PrPC expression reflects a primary mechanism of disease and is not merely a secondary consequence of other AD-associated changes. The reduction of PrPC in the brain with aging suggests that age-related decreases in PrPC may contribute to the increased incidence of AD in older people.


Subject(s)
Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , PrPC Proteins/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Female , Humans , Incidence , Male , Middle Aged , Prions/metabolism , Young Adult
9.
Int J Alzheimers Dis ; 2011: 971021, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21197404

ABSTRACT

Zinc, the most abundant trace metal in the brain, has numerous functions, both in health and in disease. Zinc is released into the synaptic cleft of glutamatergic neurons alongside glutamate from where it interacts and modulates NMDA and AMPA receptors. In addition, zinc has multifactorial functions in Alzheimer's disease (AD). Zinc is critical in the enzymatic nonamyloidogenic processing of the amyloid precursor protein (APP) and in the enzymatic degradation of the amyloid-ß (Aß) peptide. Zinc binds to Aß promoting its aggregation into neurotoxic species, and disruption of zinc homeostasis in the brain results in synaptic and memory deficits. Thus, zinc dyshomeostasis may have a critical role to play in the pathogenesis of AD, and the chelation of zinc is a potential therapeutic approach.

10.
PLoS Pathog ; 5(11): e1000666, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19936054

ABSTRACT

In prion diseases, the cellular form of the prion protein, PrP(C), undergoes a conformational conversion to the infectious isoform, PrP(Sc). PrP(C) associates with lipid rafts through its glycosyl-phosphatidylinositol (GPI) anchor and a region in its N-terminal domain which also binds to heparan sulfate proteoglycans (HSPGs). We show that heparin displaces PrP(C) from rafts and promotes its endocytosis, suggesting that heparin competes with an endogenous raft-resident HSPG for binding to PrP(C). We then utilised a transmembrane-anchored form of PrP (PrP-TM), which is targeted to rafts solely by its N-terminal domain, to show that both heparin and phosphatidylinositol-specific phospholipase C can inhibit its association with detergent-resistant rafts, implying that a GPI-anchored HSPG targets PrP(C) to rafts. Depletion of the major neuronal GPI-anchored HSPG, glypican-1, significantly reduced the raft association of PrP-TM and displaced PrP(C) from rafts, promoting its endocytosis. Glypican-1 and PrP(C) colocalised on the cell surface and both PrP(C) and PrP(Sc) co-immunoprecipitated with glypican-1. Critically, treatment of scrapie-infected N2a cells with glypican-1 siRNA significantly reduced PrP(Sc) formation. In contrast, depletion of glypican-1 did not alter the inhibitory effect of PrP(C) on the beta-secretase cleavage of the Alzheimer's amyloid precursor protein. These data indicate that glypican-1 is a novel cellular cofactor for prion conversion and we propose that it acts as a scaffold facilitating the interaction of PrP(C) and PrP(Sc) in lipid rafts.


Subject(s)
Glypicans/metabolism , Membrane Microdomains/metabolism , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Prions/metabolism , Amyloid/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Models, Animal , Heparin/pharmacology , Membrane Microdomains/drug effects , Mice , Protein Isoforms
SELECTION OF CITATIONS
SEARCH DETAIL
...