Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Neurobiol Dis ; 85: 187-205, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26563932

ABSTRACT

Accumulation of ß-amyloid (Aß) and phosphorylated tau in the brain are central events underlying Alzheimer's disease (AD) pathogenesis. Aß is generated from amyloid precursor protein (APP) by ß-site APP-cleaving enzyme 1 (BACE1) and γ-secretase-mediated cleavages. Ubiquilin-1, a ubiquitin-like protein, genetically associates with AD and affects APP trafficking, processing and degradation. Here, we have investigated ubiquilin-1 expression in human brain in relation to AD-related neurofibrillary pathology and the effects of ubiquilin-1 overexpression on BACE1, tau, neuroinflammation, and neuronal viability in vitro in co-cultures of mouse embryonic primary cortical neurons and microglial cells under acute neuroinflammation as well as neuronal cell lines, and in vivo in the brain of APdE9 transgenic mice at the early phase of the development of Aß pathology. Ubiquilin-1 expression was decreased in human temporal cortex in relation to the early stages of AD-related neurofibrillary pathology (Braak stages 0-II vs. III-IV). There was a trend towards a positive correlation between ubiquilin-1 and BACE1 protein levels. Consistent with this, ubiquilin-1 overexpression in the neuron-microglia co-cultures with or without the induction of neuroinflammation resulted in a significant increase in endogenously expressed BACE1 levels. Sustained ubiquilin-1 overexpression in the brain of APdE9 mice resulted in a moderate, but insignificant increase in endogenous BACE1 levels and activity, coinciding with increased levels of soluble Aß40 and Aß42. BACE1 levels were also significantly increased in neuronal cells co-overexpressing ubiquilin-1 and BACE1. Ubiquilin-1 overexpression led to the stabilization of BACE1 protein levels, potentially through a mechanism involving decreased degradation in the lysosomal compartment. Ubiquilin-1 overexpression did not significantly affect the neuroinflammation response, but decreased neuronal viability in the neuron-microglia co-cultures under neuroinflammation. Taken together, these results suggest that ubiquilin-1 may mechanistically participate in AD molecular pathogenesis by affecting BACE1 and thereby APP processing and Aß accumulation.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Autophagy-Related Proteins , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Survival/physiology , Coculture Techniques , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , tau Proteins/metabolism
2.
Epilepsy Res ; 112: 43-55, 2015 May.
Article in English | MEDLINE | ID: mdl-25847338

ABSTRACT

Patients with Alzheimer's disease are at increased risk for unprovoked seizures and epilepsy compared with age-matched controls. Experimental evidence suggests that neuronal hyperexcitability and epilepsy can be triggered by amyloid-ß (Aß), the main component of amyloid plaques. Previous studies demonstrated that the administration of an anticonvulsant and histone deacetylase inhibitor, valproic acid, leads to a long-lasting reduction in Aß levels. Here we used an APdE9 mouse model of Alzheimer's disease with overproduction of Aß to assess whether treatment with valproic acid initiated immediately after epilepsy onset modifies the occurrence of epileptiform activity. We also analyzed whether the effect is long-lasting and associated with antiamyloidogenesis and histone-modifications. Male APdE9 mice (15 week old) received daily intraperitoneal injections of 30mg/kg valproic acid for 1 week. After a 3-week wash-out, the same animals received injections of a higher dose of valproic acid (300mg/kg) daily for 1 week. Long-term video-electroencephalography monitoring was performed prior to, during, and after the treatments. Aß and total histone H3 and H4 acetylation levels were measured at 1 month after the final valproic acid treatment. While 30mg/kg valproic acid reduced spontaneous seizures in APdE9 mice (p<0.05, chi-square), epileptiform discharges were not reduced. Administration of 300mg/kg valproic acid, however, reduced epileptiform discharges in APdE9 mice for at least 1 week after treatment discontinuation (p<0.05, Wilcoxon test), but there was no consistent long-term effects on epileptiform activity after treatment withdrawal. Further, we found no long-lasting effect on Aß levels (p>0.05, Mann-Whitney test), only a meager increase in global acetylation of histone H3 (p<0.05), and no effects on H4 acetylation (p>0.05). In conclusion, valproic acid treatment of APdE9 mice at the stage when amyloid plaques are beginning to develop and epileptiform activity is detected reduced the amount of epileptiform activity, but the effect disappeared after treatment discontinuation.


Subject(s)
Alzheimer Disease/complications , Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Epilepsy/etiology , Valproic Acid/therapeutic use , Acetylation/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/mortality , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy/mortality , Histones/genetics , Histones/metabolism , Male , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Random Allocation , Time Factors , Video Recording
3.
Neurobiol Aging ; 36(2): 1221.e15-28, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25281018

ABSTRACT

In this study, we have assessed the expression and splicing status of genes involved in the pathogenesis or affecting the risk of Alzheimer's disease (AD) in the postmortem inferior temporal cortex samples obtained from 60 subjects with varying degree of AD-related neurofibrillary pathology. These subjects were grouped based on neurofibrillary pathology into 3 groups: Braak stages 0-II, Braak stages III-IV, and Braak stages V-VI. We also examined the right frontal cortical biopsies obtained during life from 22 patients with idiopathic shunt-responding normal pressure hydrocephalus, a disease that displays similar pathologic alterations as seen in AD. These 22 patients were categorized according to dichotomized amyloid-ß positive or negative pathology in the biopsies. We observed that the expression of FRMD4A significantly decreased, and the expression of MS4A6A significantly increased in relation to increasing AD-related neurofibrillary pathology. Moreover, the expression of 2 exons in both CLU and TREM2 significantly increased with increase in AD-related neurofibrillary pathology. However, a similar trend toward increased expression in CLU and TREM2 was observed with most of the studied exons, suggesting a global change in the expression rather than altered splicing. Correlation of gene expression with well-established AD-related factors, such as α-, ß-, and γ-secretase activities, brain amyloid-ß42 levels, and cerebrospinal fluid biomarkers, revealed a positive correlation between ß-secretase activity and the expression of TREM2 and BIN1. In expression quantitative trait loci analysis, we did not detect significant effects of the risk alleles on gene expression or splicing. Analysis of the normal pressure hydrocephalus biopsies revealed no differences in the expression or splicing profiles of the studied genes between amyloid-ß positive and negative patients. Using the protein-protein interaction-based in vitro pathway analysis tools, we found that downregulation of FRMD4A associated with increased APP-ß-secretase interaction, increased amyloid-ß40 secretion, and altered phosphorylation of tau. Taken together, our results suggest that the expression of FRMD4A, MS4A6A, CLU, and TREM2 is altered in relation to increasing AD-related neurofibrillary pathology, and that FRMD4A may play a role in amyloidogenic and tau-related pathways in AD. Therefore, investigation of gene expression changes in the brain and effects of the identified genes on disease-associated pathways in vitro may provide mechanistic insights on how alterations in these genes may contribute to AD pathogenesis.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Gene Expression Profiling , Genetic Predisposition to Disease/genetics , Transcriptome/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Brain/pathology , Female , Gene Expression , Humans , In Vitro Techniques , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurofibrils/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Risk , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism
4.
J Alzheimers Dis ; 43(2): 565-73, 2015.
Article in English | MEDLINE | ID: mdl-25096612

ABSTRACT

BACKGROUND: Several risk loci for Alzheimer's disease (AD) have been identified during recent years in large-scale genome-wide association studies. However, little is known about the mechanisms by which these loci influence AD pathogenesis. OBJECTIVE: To investigate the individual and combined risk effects of the newly identified AD loci. METHODS: Association of 12 AD risk loci with AD and AD-related cerebrospinal fluid (CSF) biomarkers was assessed. Furthermore, a polygenic risk score combining the effect sizes of the top 22 risk loci in AD was calculated for each individual among the clinical and neuropathological cohorts. Effects of individual risk loci and polygenic risk scores were assessed in relation to CSF biomarker levels as well as neurofibrillary pathology and different biochemical measures related to AD pathogenesis obtained from the temporal cortex. RESULTS: Polygenic risk scores associated with CSF amyloid-ß42 (Aß42) levels in the clinical cohort, and with soluble Aß42 levels and γ-secretase activity in the neuropathological cohort. The γ-secretase effect was independent of APOE. APOE-ε4 associated with CSF Aß42 (p < 0.001) levels. For the other risk loci, no significant associations with AD risk or CSF biomarkers were detected after multiple testing correction. CONCLUSIONS: AD risk loci polygenically contribute to Aß pathology in the CSF and temporal cortex, and this effect is potentially associated with increased γ-secretase activity.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Apolipoproteins E/genetics , Biomarkers/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Brain/metabolism , Cohort Studies , Disease Progression , Female , Genetic Testing , Genome-Wide Association Study , Genotype , Humans , Male , Neuropsychological Tests , Peptide Fragments/cerebrospinal fluid , Polymorphism, Single Nucleotide
5.
PLoS One ; 8(11): e80700, 2013.
Article in English | MEDLINE | ID: mdl-24278306

ABSTRACT

Alzheimer's disease (AD) has been postulated to involve defects in the clearance of amyloid-ß (Aß). Activation of liver X receptor α (LXRα) increases the expression of apolipoprotein E (ApoE) as well as cholesterol transporters ABCA1 and ABCG1, leading to augmented clearance of Aß. We have previously shown that the C allele of rs7120118 in the NR1H3 gene encoding LXRα reduces the risk of AD. Here, we wanted to assess whether the rs7120118 variation affects the progression of AD and modulates the expression of NR1H3 and its downstream targets APOE, ABCA1 and ABCG1.We utilized tissue samples from the inferior temporal cortex of 87 subjects, which were subdivided according to Braak staging into mild, moderate and severe AD groups on the basis of AD-related neurofibrillary pathology. APOE ε4 allele increased soluble Aß42 levels in the tissue samples in a dose-dependent manner, but did not affect the expression status of APOE. In contrast, the CC genotype of rs7120118 was underrepresented in the severe group, although this result did not reach statistical significance. Also, patients with the CC genotype of rs7120118 showed significantly decreased soluble Aß42 levels as compared to the patients with TT genotype. Although the severity of AD did not affect NR1H3 expression, the mRNA levels of NR1H3 among the patients with CT genotype of rs7120118 were significantly increased as compared to the patients with TT genotype. These results suggest that genetic variation in NR1H3 modulates the expression of LXRα and the levels of soluble Aß42.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Disease Progression , Genetic Predisposition to Disease , Orphan Nuclear Receptors/genetics , Polymorphism, Single Nucleotide/genetics , Aged, 80 and over , Alleles , Alzheimer Disease/cerebrospinal fluid , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoprotein E4/genetics , Demography , Female , Gene Expression Regulation , Humans , Liver X Receptors , Male , Orphan Nuclear Receptors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solubility , Temporal Lobe/metabolism , Temporal Lobe/pathology , tau Proteins/metabolism
6.
Biochemistry ; 52(22): 3899-912, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23663107

ABSTRACT

Ubiquilin-1 is an Alzheimer's disease-associated protein, which is known to modulate amyloid precursor protein (APP) processing, amyloid-ß (Aß) secretion, and presenilin-1 (PS1) accumulation. Here, we aim to elucidate the molecular mechanisms by which full-length transcript variant 1 of ubiquilin-1 (TV1) affects APP processing and γ-secretase function in human neuroblastoma cells stably overexpressing APP (SH-SY5Y-APP751). We found that TV1 overexpression significantly increased the level of APP intracellular domain (AICD) generation. However, there was no increase in the levels of secreted Aß40, Aß42, or total Aß, suggesting that ubiquilin-1 in particular enhances γ-secretase-mediated ε-site cleavage. This is supported by the finding that TV1 also significantly increased the level of intracellular domain generation of another γ-secretase substrate, leukocyte common antigen-related (LAR) phosphatase. However, in these cells, the increase in AICD levels was abolished, suggesting a preference of the γ-secretase for LAR over APP. TV2, another ubiquilin-1 variant that lacks the protein fragment encoded by exon 8, did not increase the level of AICD generation like TV1 did. The subcellular and plasma membrane localization of APP or γ-secretase complex components PS1 and nicastrin was not altered in TV1-overexpressing cells. Moreover, the effects of TV1 were not mediated by altered expression or APP binding of FE65, an adaptor protein thought to regulate AICD generation and stability. These data suggest that ubiquilin-1 modulates γ-secretase-mediated ε-site cleavage and thus may play a role in regulating γ-secretase cleavage of various substrates.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/drug effects , Autophagy-Related Proteins , Carrier Proteins/biosynthesis , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Humans , Peptide Fragments/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/drug effects
7.
Expert Opin Ther Targets ; 17(7): 795-810, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23600477

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is a common neurodegenerative disorder affecting an increasing number of people worldwide as the population ages. Currently, there are no drugs available that could prevent AD pathogenesis or slow down its progression. Increasing evidence links ubiquilin-1, an ubiquitin-like protein, into the pathogenic mechanisms of AD and other neurodegenerative diseases. Ubiquilin-1 has been shown to play a key role in the regulation of the levels, subcellular targeting, aggregation and degradation of various neurodegenerative disease-associated proteins. These include the amyloid precursor protein and presenilins that are intimately involved in the mechanisms of AD. AREAS COVERED: Here, the properties and diverse functions of ubiquilin-1 protein in the context of the pathogenesis of AD and other neurodegenerative disorders are discussed. This review recapitulates the available knowledge on the involvement of ubiquilin-1 in the genetic and molecular mechanisms in AD. Furthermore, the association of ubiquilin-1 with specific proteins and mechanisms involved in the pathogenesis of neurodegenerative diseases is described and the known ubiquilin-1-interacting proteins summarized. EXPERT OPINION: The variety of ubiquilin-1-interacting proteins and its central role in the regulation of protein levels and degradation provides a number of novel candidates and approaches for future research and drug discovery.


Subject(s)
Alzheimer Disease/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autophagy-Related Proteins , Humans
8.
J Alzheimers Dis ; 33(1): 23-7, 2013.
Article in English | MEDLINE | ID: mdl-22890097

ABSTRACT

Genetic variants in the granulin (GRN) gene have been shown to increase the risk of Alzheimer's disease (AD). Here, we report that the A allele of rs5848 in GRN reduces plasma granulin levels in a dose-dependent manner in a clinically-defined AD sample cohort. Similarly, the mRNA levels of granulin were decreased with respect to A allele of rs5848 in the inferior temporal cortex of neuropathologically confirmed AD patients. Our findings suggest that the A allele of rs5848 is functionally relevant by reducing the expression of granulin.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/genetics , Brain/metabolism , Brain/pathology , Down-Regulation/genetics , Genetic Variation/genetics , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Biomarkers/blood , Biomarkers/metabolism , Brain/blood supply , Cohort Studies , Female , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Male , Middle Aged , Oxidation-Reduction , Progranulins
9.
Commun Integr Biol ; 4(4): 428-32, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21966562

ABSTRACT

Controlled management of protein levels and quality is essential for normal cellular function. Specific molecular chaperones and foldases monitor the levels and assist correct folding of proteins. The ubiquitin-proteasome system recognizes and degrades misfolded proteins that can otherwise be harmful to cells. However, when misfolded or aggregated proteins excessively accumulate, they may be sequestered to the microtubule-organizing center to form aggresomes. These may then be removed from cells by autophagocytosis. Abnormal protein accumulation and aggregation is a common hallmark of many neurodegenerative diseases. In a recent study, we provide evidence that specific transcript variants (TVs) of ubiquilin-1, which are genetically and functionally associated to Alzheimer's disease (AD), regulate proteasomal and aggresomal targeting of presenilin-1 (PS1), a key player in AD pathogenesis. Our study together with current data provide interesting implications for ubiquilin-1 and its TVs in the pathogenesis of AD and other neurodegenerative diseases involving abnormal protein aggregation.

10.
Traffic ; 12(3): 330-48, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21143716

ABSTRACT

The Alzheimer's disease (AD)-associated ubiquilin-1 regulates proteasomal degradation of proteins, including presenilin (PS). PS-dependent γ-secretase generates ß-amyloid (Aß) peptides, which excessively accumulate in AD brain. Here, we have characterized the effects of naturally occurring ubiquilin-1 transcript variants (TVs) on the levels and subcellular localization of PS1 and other γ-secretase complex components and subsequent γ-secretase function in human embryonic kidney 293, human neuroblastoma SH-SY5Y and mouse primary cortical cells. Full-length ubiquilin-1 TV1 and TV3 that lacks the proteasome-interaction domain increased full-length PS1 levels as well as induced accumulation of high-molecular-weight PS1 and aggresome formation. Accumulated PS1 colocalized with TV1 or TV3 in the aggresomes. Electron microscopy indicated that aggresomes containing TV1 or TV3 were targeted to autophagosomes. TV1- and TV3-expressing cells did not accumulate other unrelated proteasome substrates, suggesting that the increase in PS1 levels was not because of a general impairment of the ubiquitin-proteasome system. Furthermore, PS1 accumulation and aggresome formation coincided with alterations in Aß levels, particularly in cells overexpressing TV3. These effects were not related to altered γ-secretase activity or PS1 binding to TV3. Collectively, our results indicate that specific ubiquilin-1 TVs can cause PS1 accumulation and aggresome formation, which may impact AD pathogenesis or susceptibility.


Subject(s)
Alzheimer Disease , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Presenilin-1/metabolism , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/metabolism , Animals , Autophagy-Related Proteins , Cells, Cultured , Gene Expression , HEK293 Cells , Humans , Mice , Mice, Transgenic , Models, Biological , Phagosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Stability
11.
Biochem Soc Trans ; 38(Pt 1): 150-5, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20074050

ABSTRACT

Abnormal protein aggregation and intracellular or extracellular accumulation of misfolded and aggregated proteins are key events in the pathogenesis of different neurodegenerative diseases. Furthermore, endoplasmic reticulum stress and impairment of the ubiquitin-proteasome system probably contribute to neurodegeneration in these diseases. A characteristic feature of AD (Alzheimer's disease) is the abnormal accumulation of Abeta (amyloid beta-peptide) in the brain. Evidence shows that the AD-associated PS (presenilin) also forms aggregates under certain conditions and that another AD-associated protein, ubiquilin-1, controls protein aggregation and deposition of aggregated proteins. Here, we review the current knowledge of ubiquilin-1 and PS in protein aggregation and related events that potentially influence neurodegeneration.


Subject(s)
Alzheimer Disease , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Autophagy-Related Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Endoplasmic Reticulum/metabolism , Humans , Inclusion Bodies/metabolism , Oxidative Stress , Presenilins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , Protein Folding
12.
J Biol Chem ; 284(49): 34433-43, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19815556

ABSTRACT

Seladin-1 is a neuroprotective protein selectively down-regulated in brain regions affected in Alzheimer disease (AD). Seladin-1 protects cells against beta-amyloid (Abeta) peptide 42- and oxidative stress-induced apoptosis activated by caspase-3, a key mediator of apoptosis. Here, we have employed RNA interference to assess the molecular effects of seladin-1 down-regulation on the beta-secretase (BACE1) function and beta-amyloid precursor protein (APP) processing in SH-SY5Y human neuroblastoma cells in both normal and apoptotic conditions. Our results show that approximately 60% reduction in seladin-1 protein levels, resembling the decrease observed in AD brain, did not significantly affect APP processing or Abeta secretion in normal growth conditions. However, under apoptosis, seladin-1 small interfering RNA (siRNA)-transfected cells showed increased caspase-3 activity on average by 2-fold when compared with control siRNA-transfected cells. Increased caspase-3 activity coincided with a significant depletion of the BACE1-sorting protein, GGA3 (Golgi-localized gamma-ear-containing ADP-ribosylation factor-binding protein), and subsequently augmented BACE1 protein levels and activity. Augmented BACE1 activity in turn correlated with the enhanced beta-amyloidogenic processing of APP and ultimately increased Abeta production. These adverse changes associated with decreased cell viability in seladin-1 siRNA-transfected cells under apoptosis. No changes in GGA3 or BACE1 levels were found after seladin-1 knockdown in normal growth conditions. Collectively, our results suggest that under stress conditions, reduced seladin-1 expression results in enhanced GGA3 depletion, which further leads to augmented post-translational stabilization of BACE1 and increased beta-amyloidogenic processing of APP. These mechanistic findings related to seladin-1 down-regulation are important in the context of AD as the oxidative stress-induced apoptosis plays a key role in the disease pathogenesis.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Amyloid Precursor Protein Secretases/biosynthesis , Apoptosis , Aspartic Acid Endopeptidases/biosynthesis , Gene Expression Regulation , Nerve Tissue Proteins/biosynthesis , Oxidoreductases Acting on CH-CH Group Donors/biosynthesis , Amyloid beta-Peptides/chemistry , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Down-Regulation , Humans , Microscopy, Confocal/methods , Models, Biological , Oxidative Stress , Protein Structure, Tertiary , Transfection
13.
Am J Med Genet B Neuropsychiatr Genet ; 150B(5): 747-50, 2009 Jul 05.
Article in English | MEDLINE | ID: mdl-19016491

ABSTRACT

Granulin protein plays an important role in neurite outgrowth and neuronal survival. Recently, it was shown that mutations in granulin (GRN) gene cause tau-negative frontotemporal dementia supporting the idea that granulin is involved in neurodegeneration. Here we have investigated whether genetic variability in the GRN gene influences also the risk of developing Alzheimer's disease (AD). Genotyping of six single nucleotide polymorphisms (SNPs) in the GRN gene among 512 AD patients and 649 control subjects originating from Finland did not show significant association with AD. However, stratification according to gender revealed a significant male-specific allele, genotype and haplotype association between AD and GRN SNPs rs4792939, rs850713, and rs5848. These data suggest that genetic variability in the GRN gene may also increase the risk for developing AD in a gender-specific manner.


Subject(s)
Alzheimer Disease/genetics , Intercellular Signaling Peptides and Proteins/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Case-Control Studies , Female , Finland , Gene Frequency , Genetics, Population , Genotype , Humans , Linkage Disequilibrium , Male , Middle Aged , Progranulins , Sex Characteristics , Young Adult
14.
Eur J Hum Genet ; 16(2): 171-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18043715

ABSTRACT

Familial early-onset Alzheimer's disease with cerebral amyloid angiopathy (EOAD/CAA) was recently associated with duplications of the gene for the amyloid-beta precursor protein (APP). In this study, we have screened for duplications of APP in patients with EOAD from Sweden and Finland. Seventy-five individuals from families with EOAD and 66 individuals with EOAD without known familial inheritance were screened by quantitative PCR. On the basis of the initial results, a portion of the samples was also investigated with quantitative multiplex PCR. No duplications of APP were identified, whereby we conclude that this is not a common cause of EOAD in the Swedish and Finnish populations, at least not in our collection of families and cases.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Gene Duplication , Adult , Age of Onset , Aged , Alzheimer Disease/complications , Alzheimer Disease/epidemiology , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy/epidemiology , Cerebral Amyloid Angiopathy/genetics , Female , Finland , Gene Dosage , Humans , Male , Middle Aged , Prevalence , Sweden
SELECTION OF CITATIONS
SEARCH DETAIL
...