Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 3(6)2020 06.
Article in English | MEDLINE | ID: mdl-32354700

ABSTRACT

Presenilins (PS) are the catalytic components of γ-secretase complexes that mediate intramembrane proteolysis. Mutations in the PS genes are a major cause of familial early-onset Alzheimer disease and affect the cleavage of the amyloid precursor protein, thereby altering the production of the amyloid ß-peptide. However, multiple additional protein substrates have been identified, suggesting pleiotropic functions of γ-secretase. Here, we demonstrate that inhibition of γ-secretase causes dysregulation of cellular lipid homeostasis, including up-regulation of liver X receptors, and complex changes in the cellular lipid composition. Genetic and pharmacological inhibition of γsecretase leads to strong accumulation of cytoplasmic lipid droplets, associated with increased levels of acylglycerols, but lowered cholesteryl esters. Furthermore, accumulation of lipid droplets was augmented by increasing levels of amyloid precursor protein C-terminal fragments, indicating a critical involvement of this γ-secretase substrate. Together, these data provide a mechanism that functionally connects γ-secretase activity to cellular lipid metabolism. These effects were also observed in human astrocytic cells, indicating an important function of γ-secretase in cells critical for lipid homeostasis in the brain.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Cholesterol Esters/metabolism , Glycerides/metabolism , Lipid Droplets/metabolism , Liver X Receptors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Line, Tumor , Diamines/pharmacology , Fibroblasts/metabolism , Gene Deletion , Homeostasis/drug effects , Homeostasis/genetics , Humans , Mice , Mice, Knockout , Presenilins/deficiency , Presenilins/genetics , Thiazoles/pharmacology , Transfection , Up-Regulation/drug effects , Up-Regulation/genetics
2.
PLoS One ; 10(11): e0143135, 2015.
Article in English | MEDLINE | ID: mdl-26606074

ABSTRACT

OBJECTIVE: This study investigated the effects of intervention with a combination of nutrients in the amyloid precursor protein-presenilin (APP-PSN) C57BL/6J double transgenic mouse model of Alzheimer's disease (AD). METHODS: A total of 72 2-month-old APP-PSN mice were randomly assigned to three groups. The model group (MG) was fed regular, unsupplemented chow, while the low- and high-dose treatment groups (LG and HG, respectively) were given a combination of nutrients that included phosphatidylserine, blueberry extracts, docosahexaenoic acid, and eicosapentaenoic acid as part of their diet. An additional 24 wild-type littermates that were fed unsupplemented chow served as the negative control group (NG). After 3 and 7 months of treatment, the cognitive performance was assessed with the Morris water maze and the shuttle box escape/avoidance task, and the biochemical parameters and oxidative stress were evaluated in both the blood and brain. RESULTS: An improvement in antioxidant capacity was observed in the treatment groups relative to the MG at 3 months, while superior behavioral test results were observed in the mice of the HG and NG groups. In the MG, pycnosis was detected in neuronal nuclei, and a loss of neurons was observed in the cerebral cortex and the hippocampus. At 7 months, the ß-amyloid1-42 peptide accumulation was significantly elevated in the MG but was markedly lower in the mice fed the nutrient combination. The antioxidant capacity and behavioral test scores were also higher in these mice. CONCLUSIONS: Early intervention with a combination of nutrients should be considered as a strategy for preventing cognitive decline and other symptoms associated with AD.


Subject(s)
Alzheimer Disease/genetics , Animal Feed , Dietary Supplements , Acetylcholine/blood , Acetylcholine/metabolism , Alzheimer Disease/diet therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal , Body Weight , Cholinesterases/blood , Cholinesterases/metabolism , Disease Models, Animal , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress , Presenilins/deficiency , Presenilins/genetics
3.
Cell Physiol Biochem ; 34(1): 92-110, 2014.
Article in English | MEDLINE | ID: mdl-24977484

ABSTRACT

BACKGROUND: Gangliosides were found to be associated with Alzheimer's disease (AD). Here we addressed a potential function of γ-secretase (presenilin) dependent cleavage of the amyloid-precursor-protein (APP) in the regulation of ganglioside de novo synthesis. METHODS: To identify a potential role of γ-secretase and APP in ganglioside de novo synthesis we used presenilin (PS) deficient and APP deficient cells and mouse brains, mutated PS as well as transgenic mice and AD post mortem brains. Changes in glucosylceramide synthase (GCS) activity were identified by incorporation of radiolabeled UDP-glucose in glucosylceramide, changes in gene expression via real-time PCR and Western blot analysis. Alterations in ganglioside levels were determined by thin layer chromatography and mass spectrometry. RESULTS: We found that PS and APP deficiency, in vitro and in vivo, resulted in increased GCS gene expression, elevated enzyme activity and thus increased glucosylceramide and total ganglioside level. Using a specific γ-secretase inhibitor revealed that PS proteolytic activity alters ganglioside homeostasis. By the use of mutated PS causing early onset AD in cell culture and transgenic mice we found that GCS is increased in AD, further substantiated by the use of AD post mortem brains, suffering from sporadic AD. CONCLUSION: APP processing regulates ganglioside de novo synthesis and is affected in AD.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/metabolism , Glucosyltransferases/metabolism , Presenilins/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Female , Gangliosides/metabolism , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Presenilins/deficiency , Presenilins/genetics , Transfection
4.
PLoS One ; 8(5): e62811, 2013.
Article in English | MEDLINE | ID: mdl-23667524

ABSTRACT

Dietary copper is essential for multicellular organisms. Copper is redox active and required as a cofactor for enzymes such as the antioxidant Superoxide Dismutase 1 (SOD1). Copper dyshomeostasis has been implicated in Alzheimer's disease. Mutations in the presenilin genes encoding PS1 and PS2 are major causes of early-onset familial Alzheimer's disease. PS1 and PS2 are required for efficient copper uptake in mammalian systems. Here we demonstrate a conserved role for presenilin in dietary copper uptake in the fly Drosophila melanogaster. Ubiquitous RNA interference-mediated knockdown of the single Drosophila presenilin (PSN) gene is lethal. However, PSN knockdown in the midgut produces viable flies. These flies have reduced copper levels and are more tolerant to excess dietary copper. Expression of a copper-responsive EYFP construct was also lower in the midgut of these larvae, indicative of reduced dietary copper uptake. SOD activity was reduced by midgut PSN knockdown, and these flies were sensitive to the superoxide-inducing chemical paraquat. These data support presenilin being needed for dietary copper uptake in the gut and so impacting on SOD activity and tolerance to oxidative stress. These results are consistent with previous studies of mammalian presenilins, supporting a conserved role for these proteins in mediating copper uptake.


Subject(s)
Copper/metabolism , Diet , Insect Proteins/metabolism , Presenilins/metabolism , Animals , Biological Transport , Conserved Sequence , Drosophila melanogaster/metabolism , Gene Knockdown Techniques , Insect Proteins/deficiency , Insect Proteins/genetics , Oxidative Stress , Presenilins/deficiency , Presenilins/genetics , RNA Interference , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
5.
Cell Death Differ ; 20(4): 639-48, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23306558

ABSTRACT

Promyelocytic leukemia (PML) is a major component of macromolecular multiprotein complexes called PML nuclear-bodies (PML-NBs). These PML-NBs recruit numerous proteins including CBP, p53 and HIPK2 in response to DNA damage, senescence and apoptosis. In this study, we investigated the effect of presenilin (PS), the main component of the γ-secretase complex, in PML/p53 expression and downstream consequences during DNA damage-induced cell death using camptothecin (CPT). We found that the loss of PS in PS knockout (KO) MEFs (mouse embryonic fibroblasts) results in severely blunted PML expression and attenuated cell death upon CPT exposure, a phenotype that is fully reversed by re-expression of PS1 in PS KO cells and recapitulated by γ-secretase inhibitors in hPS1 MEFs. Interestingly, the γ-secretase cleavage product, APP intracellular domain (AICD), together with Fe65-induced PML expression at the protein and transcriptional levels in PS KO cells. PML and p53 reciprocally positively regulated each other during CPT-induced DNA damage, both of which were dependent on PS. Finally, elevated levels of PML-NB, PML protein and PML mRNA were detected in the brain tissues from Alzheimer's disease (AD) patients, where γ-secretase activity is essential for pathogenesis. Our data provide for the first time, a critical role of the PS/AICD-PML/p53 pathway in DNA damage-induced apoptosis, and implicate this pathway in AD pathogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Apoptosis/drug effects , Camptothecin/toxicity , DNA Damage/drug effects , Nuclear Proteins/metabolism , Presenilins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Brain/metabolism , Carbamates/pharmacology , Cell Line , Dipeptides/pharmacology , Gene Expression/drug effects , Gene Knockout Techniques , HEK293 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Presenilins/deficiency , Presenilins/genetics , Promyelocytic Leukemia Protein , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Up-Regulation
6.
Article in English | MEDLINE | ID: mdl-22491782

ABSTRACT

Alzheimer's disease (AD) is a major cause of dementia in the elderly. Pathologically, AD is characterized by the accumulation of insoluble aggregates of Aß-peptides that are proteolytic cleavage products of the amyloid-ß precursor protein ("plaques") and by insoluble filaments composed of hyperphosphorylated tau protein ("tangles"). Familial forms of AD often display increased production of Aß peptides and/or altered activity of presenilins, the catalytic subunits of γ-secretase that produce Aß peptides. Although the pathogenesis of AD remains unclear, recent studies have highlighted two major themes that are likely important. First, oligomeric Aß species have strong detrimental effects on synapse function and structure, particularly on the postsynaptic side. Second, decreased presenilin function impairs synaptic transmission and promotes neurodegeneration. The mechanisms underlying these processes are beginning to be elucidated, and, although their relevance to AD remains debated, understanding these processes will likely allow new therapeutic avenues to AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Models, Biological , Presenilins/deficiency , Synapses/pathology , Synaptic Transmission/physiology , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Gene Transfer Techniques/trends , Humans , Synapses/physiology
7.
J Neurochem ; 115(5): 1215-21, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20854432

ABSTRACT

J. Neurochem. (2010) 115, 1215-1221. ABSTRACT: Synaptic dysfunction is widely thought to be a pathogenic precursor to neurodegeneration in Alzheimer's disease (AD), and the extent of synaptic loss provides the best correlate for the severity of dementia in AD patients. Presenilins 1 and 2 are the major causative genes of early-onset familial AD. Conditional inactivation of presenilins in the adult cerebral cortex results in synaptic dysfunction and memory impairment, followed by age-dependent neurodegeneration. To characterize further the consequence of presenilin inactivation in the synapse, we evaluated the temporal development of pre-synaptic and post-synaptic deficits in the Schaeffer-collateral pathway of presenilin conditional double knockout (PS cDKO) mice prior to onset of neurodegeneration. Following presenilin inactivation at 4 weeks, synaptic facilitation and probability of neurotransmitter release are impaired in PS cDKO mice at 5 weeks of age, whereas post-synaptic NMDA receptor (NMDAR)-mediated responses are normal at 5 weeks but impaired at 6 weeks of age. Long-term potentiation induced by theta burst stimulation is also reduced in PS cDKO mice at 6 weeks of age. These results show that loss of presenilins results in pre-synaptic deficits in short-term plasticity and probability of neurotransmitter release prior to post-synaptic NMDAR dysfunction, raising the possibility that presenilins may regulate post-synaptic NMDAR function in part via a trans-synaptic mechanism.


Subject(s)
Cerebral Cortex/cytology , Long-Term Potentiation/physiology , Neurons/cytology , Presenilins/deficiency , Synapses/physiology , Aging/physiology , Animals , Biophysics , Calcium/metabolism , Electric Stimulation/methods , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Presenilins/metabolism , Probability , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
8.
PLoS One ; 5(4): e10195, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20419112

ABSTRACT

Presenilins are the major causative genes of familial Alzheimer's disease (AD). Our previous study has demonstrated essential roles of presenilins in memory and neuronal survival. Here, we explore further how loss of presenilins results in age-related, progressive neurodegeneration in the adult cerebral cortex, where the pathogenesis of AD occurs. To circumvent the requirement of presenilins for embryonic development, we used presenilin conditional double knockout (Psen cDKO) mice, in which presenilin inactivation is restricted temporally and spatially to excitatory neurons of the postnatal forebrain beginning at 4 weeks of age. Increases in the number of degenerating (Fluoro-Jade B+, 7.6-fold) and apoptotic (TUNEL+, 7.4-fold) neurons, which represent approximately 0.1% of all cortical neurons, were first detected at 2 months of age when there is still no significant loss of cortical neurons and volume in Psen cDKO mice. By 4 months of age, significant loss of cortical neurons (approximately 9%) and gliosis was found in Psen cDKO mice. The apoptotic cell death is associated with caspase activation, as shown by increased numbers of cells immunoreactive for active caspases 9 and 3 in the Psen cDKO cortex. The vulnerability of cortical neurons to loss of presenilins is region-specific with cortical neurons in the lateral cortex most susceptible. Compared to the neocortex, the increase in apoptotic cell death and the extent of neurodegeneration are less dramatic in the Psen cDKO hippocampus, possibly in part due to increased neurogenesis in the aging dentate gyrus. Neurodegeneration is also accompanied with mitochondrial defects, as indicated by reduced mitochondrial density and altered mitochondrial size distribution in aging Psen cortical neurons. Together, our findings show that loss of presenilins in cortical neurons causes apoptotic cell death occurring in a very small percentage of neurons, which accumulates over time and leads to substantial loss of cortical neurons in the aging brain. The low occurrence and significant delay of apoptosis among cortical neurons lacking presenilins suggest that loss of presenilins may induce apoptotic neuronal death through disruption of cellular homeostasis rather than direct activation of apoptosis pathways.


Subject(s)
Aging , Cerebral Cortex/pathology , Nerve Degeneration/pathology , Presenilins/deficiency , Age Factors , Animals , Apoptosis , Caspases/analysis , Hippocampus , Homeostasis , Mice , Mice, Knockout , Mice, Mutant Strains , Mitochondria/pathology
9.
Genes Dev ; 23(17): 2088-101, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19723764

ABSTRACT

Canonical Notch signaling is thought to control the endocrine/exocrine decision in early pancreatic progenitors. Later, RBP-Jkappa interacts with Ptf1a and E12 to promote acinar differentiation. To examine the involvement of Notch signaling in selecting specific endocrine lineages, we deregulated this pathway by targeted deletion of presenilin1 and presenilin2, the catalytic core of gamma-secretase, in Ngn3- or Pax6-expressing endocrine progenitors. Surprisingly, whereas Pax6(+) progenitors were irreversibly committed to the endocrine fate, we discovered that Ngn3(+) progenitors were bipotential in vivo and in vitro. When presenilin amounts are limiting, Ngn3(+) progenitors default to an acinar fate; subsequently, they expand rapidly to form the bulk of the exocrine pancreas. gamma-Secretase inhibitors confirmed that enzymatic activity was required to block acinar fate selection by Ngn3 progenitors. Genetic interactions identified Notch2 as the substrate, and suggest that gamma-secretase and Notch2 act in a noncanonical titration mechanism to sequester RBP-Jkappa away from Ptf1a, thus securing selection of the endocrine fate by Ngn3 progenitors. These results revise the current view of pancreatic cell fate hierarchy, establish that Ngn3 is not in itself sufficient to commit cells to the endocrine fate in the presence of Ptf1a, reveal a noncanonical action for Notch2 protein in endocrine cell fate selection, and demonstrate that acquisition of an endocrine fate by Ngn3(+) progenitors is gamma-secretase-dependent until Pax6 expression begins.


Subject(s)
Cell Differentiation , Embryonic Development/physiology , Presenilins/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cells, Cultured , Endocrine Cells/cytology , Islets of Langerhans/cytology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Organ Size/physiology , Presenilins/deficiency
10.
J Neurosci ; 29(20): 6752-60, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19458243

ABSTRACT

The presenilin-dependent gamma-secretase processing of the beta-amyloid precursor protein (betaAPP) conditions the length of the amyloid beta peptides (Abeta) that accumulate in the senile plaques of Alzheimer's disease-affected brains. This, together with an additional presenilin-mediated epsilon-secretase cleavage, generates intracellular betaAPP-derived fragments named amyloid intracellular domains (AICDs) that regulate the transcription of several genes. We establish that presenilins control the transcription of cellular prion protein (PrP(c)) by a gamma-secretase inhibitor-sensitive and AICD-mediated process. We demonstrate that AICD-dependent control of PrP(c) involves the tumor suppressor p53. Thus, p53-deficiency abolishes the AICD-mediated control of PrP(c) transcription. Furthermore, we show that p53 directly binds to the PrP(c) promoter and increases its transactivation. Overall, our study unravels a transcriptional regulation of PrP(c) by the oncogene p53 that is directly driven by presenilin-dependent formation of AICD. Furthermore, it adds support to previous reports linking secretase activities involved in betaAPP metabolism to the physiology of PrP(c).


Subject(s)
Fibroblasts/metabolism , Presenilins/metabolism , Prions/metabolism , Tumor Suppressor Protein p53/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Cells, Cultured , Chromatin Immunoprecipitation/methods , Cyclin-Dependent Kinase Inhibitor p19/deficiency , Cyclin-Dependent Kinase Inhibitor p19/genetics , Dipeptides/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed/methods , Presenilins/deficiency , Protein Structure, Tertiary/genetics , RNA, Messenger/metabolism , Transfection/methods , Tumor Suppressor Protein p53/deficiency
11.
PLoS One ; 4(1): e4172, 2009.
Article in English | MEDLINE | ID: mdl-19137062

ABSTRACT

Presenilin (PS) is involved in many cellular events under physiological and pathological conditions. Previous reports have revealed that PS deficiency results in hyperproliferation and resistance to apoptotic cell death. In the present study, we investigated the effects of PS on beta-catenin and cell mortality during serum deprivation. Under these conditions, PS1/PS2 double-knockout MEFs showed aberrant accumulation of phospho-beta-catenin, higher ROS generation, and notable cell death. Inhibition of beta-catenin phosphorylation by LiCl reversed ROS generation and cell death in PS deficient cells. In addition, the K19/49R mutant form of beta-catenin, which undergoes normal phosphorylation but not ubiquitination, induced cytotoxicity, while the phosphorylation deficient S37A beta-catenin mutant failed to induce cytotoxicity. These results indicate that aberrant accumulation of phospho-beta-catenin underlies ROS-mediated cell death in the absence of PS. We propose that the regulation of beta-catenin is useful for identifying therapeutic targets of hyperproliferative diseases and other degenerative conditions.


Subject(s)
Cell Death/physiology , Cell Line/metabolism , Presenilins/deficiency , Reactive Oxygen Species/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Phosphorylation , Presenilins/genetics , Presenilins/metabolism
12.
Nat Med ; 14(3): 299-305, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18297084

ABSTRACT

Notch signaling is a key mechanism in the control of embryogenesis. However, its in vivo function during mesenchymal cell differentiation, and, specifically, in bone homeostasis, remains largely unknown. Here, we show that osteoblast-specific gain of Notch function causes severe osteosclerosis owing to increased proliferation of immature osteoblasts. Under these pathological conditions, Notch stimulates early osteoblastic proliferation by upregulating the genes encoding cyclin D, cyclin E and Sp7 (osterix). The intracellular domain of Notch1 also regulates terminal osteoblastic differentiation by directly binding Runx2 and repressing its transactivation function. In contrast, loss of all Notch signaling in osteoblasts, generated by deletion of the genes encoding presenilin-1 and presenilin-2 in bone, is associated with late-onset, age-related osteoporosis, which in turn results from increased osteoblast-dependent osteoclastic activity due to decreased osteoprotegerin mRNA expression in these cells. Together, these findings highlight the potential dimorphic effects of Notch signaling in bone homeostasis and may provide direction for novel therapeutic applications.


Subject(s)
Bone and Bones/metabolism , Homeostasis/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Bone and Bones/cytology , Cell Line , Cell Proliferation , Humans , Mice , Mice, Transgenic , Models, Biological , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteoporosis/metabolism , Osteosclerosis/genetics , Osteosclerosis/metabolism , Presenilins/deficiency , Receptors, Notch/genetics , Transcription Factors , Transfection
13.
J Neurochem ; 104(3): 683-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18005001

ABSTRACT

Sequential cleavages of the beta-amyloid precursor protein cleaving enzyme 1 (BACE1) by beta-secretase and gamma-secretase generate the amyloid beta-peptides, believed to be responsible of synaptic dysfunction and neuronal cell death in Alzheimer's disease (AD). Levels of BACE1 are increased in vulnerable regions of the AD brain, but the underlying mechanism is unknown. Here we show that oxidative stress (OS) stimulates BACE1 expression by a mechanism requiring gamma-secretase activity involving the c-jun N-terminal kinase (JNK)/c-jun pathway. BACE1 levels are increased in response to OS in normal cells, but not in cells lacking presenilins or amyloid precursor protein. Moreover, BACE1 is induced in association with OS in the brains of mice subjected to cerebral ischaemia/reperfusion. The OS-induced BACE1 expression correlates with an activation of JNK and c-jun, but is absent in cultured cells or mice lacking JNK. Our findings suggest a mechanism by which OS induces BACE1 transcription, thereby promoting production of pathological levels of amyloid beta in AD.


Subject(s)
Amyloid Precursor Protein Secretases/pharmacology , Amyloid beta-Protein Precursor/metabolism , Oxidative Stress/physiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/drug effects , Animals , Aspartic Acid Endopeptidases/metabolism , Cells, Cultured , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Feedback/drug effects , Feedback/physiology , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Infarction, Middle Cerebral Artery/physiopathology , MAP Kinase Kinase 4/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Presenilins/deficiency , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors , Transfection/methods
14.
J Exp Med ; 204(9): 2115-29, 2007 Sep 03.
Article in English | MEDLINE | ID: mdl-17698590

ABSTRACT

TCRalphabeta signaling is crucial for the maturation of CD4 and CD8 T cells, but the role of the Notch signaling pathway in this process is poorly understood. Genes encoding Presenilin (PS) 1/2 were deleted to prevent activation of the multiple Notch receptors expressed by developing thymocytes. PS1/2 knockout thymocyte precursors inefficiently generate CD4 T cells, a phenotype that is most pronounced when thymocytes bear a single major histocompatibility complex (MHC) class II-restricted T cell receptor (TCR). Diminished T cell production correlated with evidence of impaired TCR signaling, and could be rescued by manipulations that enhance MHC recognition. Although Notch appears to directly regulate binary fate decisions in many systems, these findings suggest a model in which PS-dependent Notch signaling influences positive selection and the development of alphabeta T cells by modifying TCR signal transduction.


Subject(s)
Presenilins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Signal Transduction , T-Lymphocytes/cytology , Alleles , Animals , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , CD4 Antigens/immunology , CD5 Antigens/genetics , Calcium/metabolism , Cross-Linking Reagents , Gene Deletion , Histocompatibility Antigens/immunology , Integrases/metabolism , Lectins, C-Type , Ligands , Mice , Mice, Knockout , Presenilins/deficiency , Thymus Gland/cytology , Transgenes , Up-Regulation/genetics
15.
Dev Neurobiol ; 67(12): 1598-613, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17562530

ABSTRACT

Alzheimer's disease is a neurodegenerative disorder characterized by progressive memory and cognitive decline that is associated with changes in synaptic plasticity and neuronal cell loss. Recent evidence suggests that some of these defects may be due to a loss of normal presenilin activity. Here, we have examined the effect of loss of Drosophila presenilin (psn) function on synaptic plasticity and learning. Basal transmitter release was elevated in psn mutants while both paired pulse synaptic plasticity and post-tetanic potentiation were impaired. These defects in synaptic strength and plasticity were not due to developmental defects in NMJ morphology. We also found that psn null terminals take up significantly less FM 4-64 than control terminals when loaded with high frequency stimulation, suggesting a defect in synaptic vesicle availability or mobilization. To determine whether these reductions in synaptic plasticity had any impact on learning, we tested the larvae for defects in associative learning. Using both olfactory and visual learning assays, we found that associative learning is impaired in psn mutants compared with controls. Both the learning and synaptic defects could be rescued by expression of a full length psn transgene suggesting the defects are specifically due to a loss of psn function. Taken together, these results provide the first evidence of learning and synaptic defects in a Drosophila psn mutant and strongly suggest a presynaptic role for presenilin in normal neuronal function.


Subject(s)
Association Learning/physiology , Neuronal Plasticity/physiology , Presenilins/deficiency , Presynaptic Terminals/metabolism , Synaptic Transmission/physiology , Action Potentials/physiology , Animals , Animals, Genetically Modified , Blotting, Western , Drosophila , Electrophysiology , Female , Gene Expression , Immunohistochemistry , Male , Mutation , Neuromuscular Junction/anatomy & histology , Neuromuscular Junction/physiology , Neurotransmitter Agents/metabolism , Presenilins/genetics , Synaptic Vesicles/metabolism
17.
Proc Natl Acad Sci U S A ; 104(2): 403-9, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17197420

ABSTRACT

Dominantly inherited mutations in the genes encoding presenilins (PS) and the amyloid precursor protein (APP) are the major causes of familial Alzheimer's disease (AD). The prevailing view of AD pathogenesis posits that accumulation of beta-amyloid (Abeta) peptides, particularly Abeta42, is the central event triggering neurodegeneration. Emerging evidence, however, suggests that loss of essential functions of PS could better explain dementia and neurodegeneration in AD. First, conditional inactivation of PS in the adult mouse brain causes progressive memory loss and neurodegeneration resembling AD, whereas mouse models based on overproduction of Abeta have failed to produce neurodegeneration. Second, whereas pathogenic PS mutations enhance Abeta42 production, they typically reduce Abeta40 generation and impair other PS-dependent activities. Third, gamma-secretase inhibitors can enhance the production of Abeta42 while blocking other gamma-secretase activities, thus mimicking the effects of PS mutations. Finally, PS mutations have been identified in frontotemporal dementia, which lacks amyloid pathology. Based on these and other observations, we propose that partial loss of PS function may underlie memory impairment and neurodegeneration in the pathogenesis of AD. We also speculate that Abeta42 may act primarily to antagonize PS-dependent functions, possibly by operating as an active site-directed inhibitor of gamma-secretase.


Subject(s)
Alzheimer Disease/etiology , Presenilins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Models, Neurological , Molecular Sequence Data , Mutation , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Presenilins/deficiency , Presenilins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...