Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Neural Transm (Vienna) ; 108(10): 1175-80, 2001.
Article in English | MEDLINE | ID: mdl-11725820

ABSTRACT

In order to identify gene variants related to the serotonergic neurotransmitter system that possibly represent a hereditary risk factor for sporadic Alzheimer's disease (AD), patients suffering from AD and non-demented psychiatric inpatients without symptoms of dementia were genotyped for polymorphisms of HTR6 (267C/T) and HTR2A (-1438G/A). Although there was a tendency toward an increased number of the genotype TT of the 5-HT6 receptor polymorphism in AD patients when compared to controls (2.8% vs. 1.3%), neither this nor the 5-HT2A promoter polymorphism showed significant differences in their genotypic or allelic distribution among patients and controls. These polymorphisms probably do not represent major genetic risk factors of AD. However, further studies including other genetic variants of the serotonergic neurotransmitter system are needed in order to elucidate their role in AD.


Subject(s)
Alzheimer Disease/genetics , Polymorphism, Genetic/genetics , Receptors, Serotonin/genetics , Alleles , Chi-Square Distribution , Genetic Variation/genetics , Genotype , Humans , Promoter Regions, Genetic/genetics , Receptor, Serotonin, 5-HT2A
2.
Biol Psychiatry ; 50(10): 809-12, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11720700

ABSTRACT

BACKGROUND: In view of the effects of stress on synaptic plasticity, the regulation of synaptophysin and synaptotagmin expression by immobilization was analyzed by in situ hybridization. METHODS: Rats were exposed to immobilization stress, which induced typical behavioral alterations, such as reduced locomotor activity after stress exposure. Determination of mRNA levels of the integral synaptic vesicle proteins was performed immediately after acute or chronic immobilization. RESULTS: The results demonstrate that stress exposure leads to reduced expression of synaptophysin but increased expression of synaptotagmin in the hippocampus. CONCLUSIONS: This rapid and differential regulation of synaptic vesicle proteins could be responsible for some of the morphological, biochemical, and behavioral changes observed after stress exposure. These changes may be relevant to such clinical disorders as psychoses, depression, and posttraumatic stress disorder that are sensitive to stress and involve changes in neural and synaptic plasticity.


Subject(s)
Calcium-Binding Proteins , Hippocampus/pathology , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Stress, Psychological/complications , Synaptophysin/genetics , Animals , Gene Expression Regulation/physiology , Neuronal Plasticity/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Restraint, Physical , Stress, Psychological/pathology , Synaptotagmins
3.
J Biol Chem ; 276(10): 7233-9, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11084029

ABSTRACT

Familial Alzheimer's disease (FAD) is frequently associated with mutations in the presenilin-1 (PS1) gene. Almost all PS1-associated FAD mutations reported so far are exchanges of single conserved amino acids and cause the increased production of the highly amyloidogenic 42-residue amyloid beta-peptide Abeta42. Here we report the identification and pathological function of an unusual FAD-associated PS1 deletion (PS1 DeltaI83/DeltaM84). This FAD mutation is associated with spastic paraparesis clinically and causes accumulation of noncongophilic Abeta-positive "cotton wool" plaques in brain parenchyma. Cerebral amyloid angiopathy due to Abeta deposition was widespread as were neurofibrillary tangles and neuropil threads, although tau-positive neurites were sparse. Although significant deposition of Abeta42 was observed, no neuritic pathology was associated with these unusual lesions. Overexpressing PS1 DeltaI83/DeltaM84 in cultured cells results in a significantly elevated level of the highly amyloidogenic 42-amino acid amyloid beta-peptide Abeta42. Moreover, functional analysis in Caenorhabditis elegans reveals reduced activity of PS1 DeltaI83/DeltaM84 in Notch signaling. Our data therefore demonstrate that a small deletion of PS proteins can pathologically affect PS function in endoproteolysis of beta-amyloid precursor protein and in Notch signaling. Therefore, the PS1 DeltaI83/DeltaM84 deletion shows a very similar biochemical/functional phenotype like all other FAD-associated PS1 or PS2 point mutations. Since increased Abeta42 production is not associated with classical senile plaque formation, these data demonstrate that amyloid plaque formation is not a prerequisite for dementia and neurodegeneration.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Gene Deletion , Membrane Proteins/genetics , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Plaque, Amyloid/chemistry , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Line , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , DNA, Complementary/metabolism , Female , Flavin-Adenine Dinucleotide/genetics , Humans , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mutation , Pedigree , Phenotype , Point Mutation , Precipitin Tests , Presenilin-1 , Receptors, Notch , Signal Transduction
4.
Nat Cell Biol ; 2(11): 848-51, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11056541

ABSTRACT

Endoproteolysis of beta-amyloid precursor protein (betaAPP) and Notch requires conserved aspartate residues in presenilins 1 and 2 (PS1 and PS2). Although PS1 and PS2 have therefore been proposed to be aspartyl proteases, no homology to other aspartyl proteases has been found. Here we identify homology between the presenilin active site and polytopic aspartyl proteases of bacterial origin, thus supporting the hypothesis that presenilins are novel aspartyl proteases.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Endopeptidases , Glycine/metabolism , Membrane Proteins/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/physiology , Bacteria/enzymology , Bacterial Proteins/metabolism , Cell Line , Conserved Sequence , Glycine/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Mutagenesis, Site-Directed , Peptide Fragments/metabolism , Presenilin-1 , Presenilin-2 , Receptors, Notch
5.
Neurosci Lett ; 294(1): 63-5, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11044587

ABSTRACT

The role of the deletion/insertion polymorphism within the promoter region of the serotonin transporter gene (5-HTT) is under discussion as a potential genetic risk factor for Alzheimers's disease (AD). Here we report significant differences in the allelic distribution of this polymorphism with a higher frequency of the short variant allele in AD patients when compared to controls. This difference was independent of the apolipoproteinE genotype. Thus, our study supports the notion that genetic alterations in the serontonergic neurotransmitter system may be involved in the etiopathogenesis of AD. However, given the reported negative findings, we are presently trying to identify diagnostic subgroups for which the 5-HTT promoter polymorphism represents a susceptibility locus.


Subject(s)
Alzheimer Disease/genetics , Carrier Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Transport Proteins , Nerve Tissue Proteins , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Alleles , Alzheimer Disease/epidemiology , Apolipoprotein E4 , Apolipoproteins E/genetics , Female , Gene Frequency , Genetic Carrier Screening , Genetic Predisposition to Disease , Genetic Testing , Humans , Male , Prevalence , Risk Factors , Serotonin Plasma Membrane Transport Proteins , White People/genetics
6.
FEBS Lett ; 463(3): 245-9, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10606730

ABSTRACT

Proteases not only play a fundamental role in numerous physiological processes, but are also involved in several human diseases including Alzheimer's disease (AD). A key protease implicated in AD is the so far unidentified gamma-secretase, which cleaves the membrane-bound beta-amyloid precursor protein (betaAPP) at the C-terminus of its amyloid domain within the membrane to release the neurotoxic amyloid beta-peptide. In order to allow the isolation of proteases, which specifically cleave membrane-bound substrates within or in the vicinity of a transmembrane domain, we developed a reporter gene assay in Saccharomyces cerevisiae. This assay may allow the identification of genes encoding target proteases that specifically cleave membrane bound substrates by transforming expression libraries.


Subject(s)
Endopeptidases/analysis , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Binding Sites , Caspase 3 , Caspases/genetics , DNA-Binding Proteins , Endopeptidases/genetics , Endopeptidases/metabolism , Fungal Proteins/genetics , Genes, Reporter , Membrane Proteins/analysis , Plasmids , Proteins/genetics , Receptors, Notch , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Transcription Factors/genetics
7.
Biochemistry ; 38(44): 14600-5, 1999 Nov 02.
Article in English | MEDLINE | ID: mdl-10545183

ABSTRACT

Alzheimer's disease (AD) is characterized by the invariant accumulation of senile plaques predominantly composed of the pathologically relevant 42-amino acid amyloid beta-peptide (Abeta42). The presenilin (PS) proteins play a key role in Abeta generation. FAD-associated mutations in PS1 and PS2 enhance the production of Abeta42, and PS1 is required for physiological Abeta production, since a gene knockout of PS1 and dominant negative mutations of PS1 abolish Abeta generation. PS proteins undergo endoproteolytic processing, and current evidence indicates that fragment formation may be required for the amyloidogenic function of PS. We have now determined the sequence requirements for endoproteolysis of PS1. Mutagenizing amino acids at the previously determined major cleavage site (amino acid 298) had no effect on PS1 endoproteolysis. In contrast, mutations or deletions at the additional cleavage site around amino acid 292 blocked endoproteolysis. The uncleavable PS1 derivatives accumulated as full-length proteins and replaced the endogenous PS1 proteins. In contrast to the previously described aspartate mutations within transmembrane domains 6 and 7, the uncleaved PS1 variants do not act as dominant negative inhibitors of Abeta production. Moreover, when a FAD-associated mutation (M146L) was combined with a mutation blocking endoproteolysis, Abeta42 production still reached pathological levels. These data therefore demonstrate that endoproteolysis of presenilins is not an absolute prerequisite for the amyloidogenic function of PS1. These data also show that accumulation of the PS1 holoprotein is not associated with the pathological activity of PS1 mutations as suggested previously.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Amino Acid Sequence , Binding Sites/genetics , Cell Line , DNA, Complementary/genetics , Endopeptidases/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , Point Mutation , Presenilin-1 , Transfection
8.
J Biol Chem ; 274(40): 28669-73, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10497236

ABSTRACT

Presenilin-1 (PS1) facilitates gamma-secretase cleavage of the beta-amyloid precursor protein and the intramembraneous cleavage of Notch1. Although Alzheimer's disease-associated mutations in the homologous presenilin (PS2) gene elevate amyloid beta-peptide (Abeta42) production like PS1 mutations, here we demonstrate that a gene ablation of PS2 (unlike that of PS1) in mice does not result in a severe phenotype resembling that of Notch-ablated animals. To investigate the amyloidogenic function of PS2 more directly, we mutagenized a conserved aspartate at position 366 to alanine, because the corresponding residue of PS1 is known to be required for its amyloidogenic function. Cells expressing the PS2 D366A mutation exhibit significant deficits in proteolytic processing of beta-amyloid precursor protein indicating a defect in gamma-secretase activity. The reduced gamma-secretase activity results in the almost complete inhibition of Abeta and p3 production in cells stably expressing PS2 D366A, whereas cells overexpressing the wild-type PS2 cDNA produce robust levels of Abeta and p3. Using highly sensitive in vivo assays, we demonstrate that the PS2 D366A mutation not only blocks gamma-secretase activity but also inactivates PS2 activity in Notch signaling by inhibiting the proteolytic release of the cytoplasmic Notch1 domain. These data suggest that PS2 is functionally involved in Abeta production and Notch signaling by facilitating similar proteolytic cleavages.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Peptide Fragments/antagonists & inhibitors , Signal Transduction/genetics , Amyloid beta-Peptides/biosynthesis , Animals , Animals, Genetically Modified , Cell Line , Humans , Hydrolysis , Membrane Proteins/physiology , Mice , Mice, Knockout , Peptide Fragments/biosynthesis , Presenilin-2 , Receptors, Notch
9.
J Biol Chem ; 274(12): 7615-8, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075646

ABSTRACT

The two homologous presenilins are key factors for the generation of amyloid beta-peptide (Abeta), since Alzheimer's disease (AD)-associated mutations enhance the production of the pathologically relevant 42-amino acid Abeta (Abeta42), and a gene knockout of presenilin-1 (PS1) significantly inhibits total Abeta production. Presenilins undergo proteolytic processing within the domain encoded by exon 9, a process that may be closely related to their biological and pathological activity. An AD-associated mutation within the PS1 gene deletes exon 9 (PS1Deltaexon9) due to a splicing error and results in the accumulation of the uncleaved full-length protein. We now demonstrate the unexpected finding that the pathological activity of PS1Deltaexon9 is independent of its lack to undergo proteolytic processing, but is rather due to a point mutation (S290C) occurring at the aberrant exon 8/10 splice junction. Mutagenizing the cysteine residue at position 290 to the original serine residue completely inhibits the pathological activity in regard to the elevated production of Abeta42. Like PS1Deltaexon9, the resulting presenilin variant (PS1Deltaexon9 C290S) accumulates as an uncleaved protein and fully replaces endogenous presenilin fragments. Moreover, PS1Deltaexon9 C290S exhibits a significantly increased biological activity in a highly sensitive in vivo assay as compared with the AD-associated mutation. Therefore not only the increased Abeta42 production but also the decreased biological function of PS1Deltaexon9 is due to a point mutation and independent of the lack of proteolytic processing.


Subject(s)
Alzheimer Disease/genetics , Exons , Membrane Proteins/genetics , Point Mutation , Animals , Animals, Genetically Modified , Cell Line , Humans , Presenilin-1 , Protein Processing, Post-Translational , RNA Splicing
10.
J Biol Chem ; 273(48): 32322-31, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9822712

ABSTRACT

Numerous mutations causing early onset Alzheimer's disease have been identified in the presenilin (PS) genes, particularly the PS1 gene. Like the mutations identified within the beta-amyloid precursor protein gene, PS mutations cause the increased generation of a highly neurotoxic variant of amyloid beta-peptide. PS proteins are proteolytically processed to an N-terminal approximately 30-kDa (NTF) and a C-terminal approximately 20-kDa fragment (CTF20) that form a heterodimeric complex. We demonstrate that this complex is resistant to proteolytic degradation, whereas the full-length precursor is rapidly degraded. Degradation of the PS1 holoprotein is sensitive to inhibitors of the proteasome. Formation of a heterodimeric complex is required for the stability of both PS1 fragments, since fragments that do not co-immunoprecipitate with the PS complex are rapidly degraded by the proteasome. Mutant PS fragments not incorporated into the heterodimeric complex lose their pathological activity in abnormal amyloid beta-peptide generation even after inhibition of their proteolytic degradation. The PS1 heterodimeric complex can be attacked by proteinases of the caspase superfamily that generate an approximately 10-kDa proteolytic fragment (CTF10) from CTF20. CTF10 is rapidly degraded most likely by a calpain-like cysteine proteinase. From these data we conclude that PS1 metabolism is highly controlled by multiple proteolytic activities indicating that subtle changes in fragment generation/degradation might be important for Alzheimer's disease-associated pathology.


Subject(s)
Cysteine Endopeptidases/metabolism , Gene Expression Regulation , Membrane Proteins/genetics , Multienzyme Complexes/metabolism , Protein Processing, Post-Translational , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Cell Line , Dimerization , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Models, Molecular , Peptide Fragments/chemistry , Presenilin-1 , Proteasome Endopeptidase Complex , Protein Structure, Secondary , Transfection
11.
J Biol Chem ; 273(6): 3205-11, 1998 Feb 06.
Article in English | MEDLINE | ID: mdl-9452432

ABSTRACT

Mutations in the presenilin (PS) genes are linked to early onset familial Alzheimer's disease (FAD). PS-1 proteins are proteolytically processed by an unknown protease to two stable fragments of approximately 30 kDa (N-terminal fragment (NTF)) and approximately 20 kDa (C-terminal fragment (CTF)) (Thinakaran, G., Borchelt, D. R., Lee, M. K., Slunt, H. H., Spitzer, L., Kim, G., Ratovitsky, T., Davenport, F., Nordstedt, C., Seeger, M., Hardy, J., Levey, A. I., Gandy, S. E., Jenkins, N. A., Copeland, N. G., Price, D. L., and Sisodia, S. S. (1996) Neuron 17, 181-190). Here we show that the CTF and NTF of PS-1 bind to each other. Fractionating proteins from 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonic acid-extracted membrane preparations by velocity sedimentation reveal a high molecular mass SDS and Triton X-100-sensitive complex of approximately 100-150 kDa. To prove if both proteolytic fragments of PS-1 are bound to the same complex, we performed co-immunoprecipitations using multiple antibodies specific to the CTF and NTF of PS-1. These experiments revealed that both fragments of PS-1 occur as a tightly bound non-covalent complex. Upon overexpression, unclipped wild type PS-1 sediments at a lower molecular weight in glycerol velocity gradients than the endogenous fragments. In contrast, the non-cleavable, FAD-associated PS-1 Deltaexon 9 sediments at a molecular weight similar to that observed for the endogenous proteolytic fragments. This result may indicate that the Deltaexon 9 mutation generates a mutant protein that exhibits biophysical properties similar to the naturally occurring PS-1 fragments. This could explain the surprising finding that the Deltaexon 9 mutation is functionally active, although it cannot be proteolytically processed (Baumeister, R., Leimer, U., Zweckbronner, I., Jakubek, C., Grünberg, J., and Haass, C. (1997) Genes & Function 1, 149-159; Levitan, D., Doyle, T., Brousseau, D., Lee, M., Thinakaran, G., Slunt, H., Sisodia, S., and Greenwald, I. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 14940-14944). Formation of a high molecular weight complex of PS-1 composed of both endogenous PS-1 fragments may also explain the recent finding that FAD-associated mutations within the N-terminal portion of PS-1 result in the hyperaccumulation not only of the NTF but also of the CTF (Lee, M. K., Borchelt, D. R., Kim, G., Thinakaran, G., Slunt, H. H., Ratovitski, T., Martin, L. J., Kittur, A., Gandy, S., Levey, A. I., Jenkins, N., Copeland, N., Price, D. L., and Sisodia, S. S. (1997) Nat. Med. 3, 756-760). Moreover, these results provide a model to understand the highly regulated expression and processing of PS proteins.


Subject(s)
Alzheimer Disease/metabolism , Membrane Proteins/metabolism , Cell Line , Dimerization , Exons , Humans , Hydrolysis , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Weight , Peptide Fragments , Precipitin Tests , Presenilin-1 , Sequence Deletion
12.
Proc Natl Acad Sci U S A ; 94(10): 5349-54, 1997 May 13.
Article in English | MEDLINE | ID: mdl-9144240

ABSTRACT

The majority of familial Alzheimer disease mutations are linked to the recently cloned presenilin (PS) genes, which encode two highly homologous proteins (PS-1 and PS-2). It was shown that the full-length PS-2 protein is phosphorylated constitutively within its N-terminal domain by casein kinases, whereas the PS-1 protein is not. Full-length PS proteins undergo endoproteolytic cleavage within their hydrophilic loop domain resulting in the formation of approximately 20-kDa C-terminal fragments (CTF) and approximately 30-kDa N-terminal fragments [Thinakaran, G., et al. (1996) Neuron 17, 181-190]. Here we describe the surprising finding that the CTF of PS-1 is phosphorylated by protein kinase C (PKC). Stimulation of PKC causes a 4- to 5-fold increase of the phosphorylation of the approximately 20-kDa CTF of PS-1 resulting in reduced mobility in SDS gels. PKC-stimulated phosphorylation occurs predominantly on serine residues and can be induced either by direct stimulation of PKC with phorbol-12,13-dibutyrate or by activation of the m1 acetylcholine receptor-signaling pathway with the muscarinic agonist carbachol. However, phosphorylation of full-length PS-1 and PS-2 is not altered upon PKC stimulation. In addition, a mutant form of PS-1 lacking exon 10, which does not undergo endoproteolytic cleavage [Thinakaran, G., et al. (1996) Neuron 17, 181-190] is not phosphorylated by PKC, although it still contains all PKC phosphorylation sites conserved between different species. These results show that PKC phosphorylates the PS-1 CTF. Therefore, endoproteolytic cleavage of full-length PS-1 results in the generation of an in vivo substrate for PKC. The selective phosphorylation of the PS-1 CTF indicates that the physiological and/or pathological properties of the CTF are regulated by PKC activity.


Subject(s)
Alzheimer Disease/metabolism , Membrane Proteins/metabolism , Protein Kinase C/metabolism , Protein Processing, Post-Translational , Alkaline Phosphatase , Alzheimer Disease/genetics , Amino Acid Sequence , Base Sequence , Cell Line , Conserved Sequence , DNA Primers , Humans , Kidney , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Presenilin-1 , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Transfection
13.
Mol Med ; 2(6): 673-91, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8972483

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the deposition of extracellular senile plaques composed of amyloid beta-peptide (A beta). Whereas most cases of AD occur sporadically, about 10% of AD cases are inherited as a fully penetrant autosomal dominant trait. Mutations in the recently cloned Presenilin genes (PS-1 and PS-2) are by far the most common cause of early onset familial AD. MATERIALS AND METHODS: Cellular expression of endogenous and overexpressed PS proteins was analyzed by immunocytochemistry and metabolic labeling followed by immunoprecipitation. In vivo phosphorylation sites of PS proteins were analyzed by extensive mutagenesis. RESULTS: PS-1 as well as PS-2 proteins were localized predominantly within the endoplasmic reticulum (ER). However, small amounts of the PS proteins were detected within the Golgi compartment, where they colocalize with the beta-amyloid precursor protein (beta APP). The PS-2 protein was found to be highly phosphorylated, whereas very little phosphorylation was observed for PS-1. The selective phosphorylation of PS-2 occurs exclusively on serine residues. In vivo phosphorylation of PS-2 was mapped to serine residues 7, 9, and 19 within an acidic stretch at the N terminus, which is absent in PS-1. casein kinase (CK)-1 and CK-2 were shown to phosphorylate the N terminus of PS-2 in vitro. CONCLUSIONS: The majority of PS proteins were detected in the ER where little if any proteolytic processing of beta APP was reported. ER retention of PS proteins might occur by intramolecular aggregation. Small amounts of PS proteins were also detected in the Golgi where they colocalized with beta APP. This might suggest that potential interactions between PS proteins and beta APP could occur within the Golgi. Selective phosphorylation of PS-2 proteins within the acidic domain missing in PS-1 indicates differences in the biological functions and regulation of the two highly homologous proteins.


Subject(s)
Alzheimer Disease/metabolism , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Amino Acid Sequence , Amyloid beta-Protein Precursor/metabolism , Animals , Blotting, Western , COS Cells , DNA Primers , Endoplasmic Reticulum/chemistry , Gene Expression Regulation/genetics , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Humans , Immunohistochemistry , Membrane Proteins/genetics , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Phosphorylation , Presenilin-1 , Presenilin-2 , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Processing, Post-Translational/genetics , Sequence Alignment , Transfection/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...