Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 16: 1279740, 2023.
Article in English | MEDLINE | ID: mdl-37908487

ABSTRACT

Recent clinical data with three therapeutic anti-Aß antibodies have demonstrated that removal of Aß-amyloid plaques in early Alzheimer's disease (AD) can attenuate disease progression. This ground-breaking progress in AD medicine has validated both the amyloid cascade hypothesis and Aß-amyloid as therapeutic targets. These results also strongly support therapeutic approaches that aim to reduce the production of amyloidogenic Aß to prevent the formation of Aß-pathology. One such strategy, so-called gamma-secretase modulators (GSM), has been thoroughly explored in preclinical settings but has yet to be fully tested in clinical trials. Recent scientific progress has shed new light on the role of Aß in Alzheimer's disease and suggests that GSMs exhibit specific pharmacological features that hold great promise for the prevention and treatment of Alzheimer's disease. In this short review, we discuss the data that support why it is important to continue to progress in this class of compounds.

2.
Nat Commun ; 14(1): 5804, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37726325

ABSTRACT

Prosaposin (PSAP) modulates glycosphingolipid metabolism and variants have been linked to Parkinson's disease (PD). Here, we find altered PSAP levels in the plasma, CSF and post-mortem brain of PD patients. Altered plasma and CSF PSAP levels correlate with PD-related motor impairments. Dopaminergic PSAP-deficient (cPSAPDAT) mice display hypolocomotion and depression/anxiety-like symptoms with mildly impaired dopaminergic neurotransmission, while serotonergic PSAP-deficient (cPSAPSERT) mice behave normally. Spatial lipidomics revealed an accumulation of highly unsaturated and shortened lipids and reduction of sphingolipids throughout the brains of cPSAPDAT mice. The overexpression of α-synuclein via AAV lead to more severe dopaminergic degeneration and higher p-Ser129 α-synuclein levels in cPSAPDAT mice compared to WT mice. Overexpression of PSAP via AAV and encapsulated cell biodelivery protected against 6-OHDA and α-synuclein toxicity in wild-type rodents. Thus, these findings suggest PSAP may maintain dopaminergic lipid homeostasis, which is dysregulated in PD, and counteract experimental parkinsonism.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Mice , alpha-Synuclein/genetics , Dopamine , Dopaminergic Neurons , Parkinson Disease/genetics , Saposins/genetics , Sphingolipids
3.
EMBO Mol Med ; 15(2): e16556, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36524456

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic form of familial small vessel disease; no preventive or curative therapy is available. CADASIL is caused by mutations in the NOTCH3 gene, resulting in a mutated NOTCH3 receptor, with aggregation of the NOTCH3 extracellular domain (ECD) around vascular smooth muscle cells. In this study, we have developed a novel active immunization therapy specifically targeting CADASIL-like aggregated NOTCH3 ECD. Immunizing CADASIL TgN3R182C150 mice with aggregates composed of CADASIL-R133C mutated and wild-type EGF1-5 repeats for a total of 4 months resulted in a marked reduction (38-48%) in NOTCH3 deposition around brain capillaries, increased microglia activation and lowered serum levels of NOTCH3 ECD. Active immunization did not impact body weight, general behavior, the number and integrity of vascular smooth muscle cells in the retina, neuronal survival, or inflammation or the renal system, suggesting that the therapy is tolerable. This is the first therapeutic study reporting a successful reduction of NOTCH3 accumulation in a CADASIL mouse model supporting further development towards clinical application for the benefit of CADASIL patients.


Subject(s)
CADASIL , Animals , Mice , Brain/metabolism , CADASIL/genetics , CADASIL/therapy , Capillaries/metabolism , Disease Models, Animal , Immunotherapy, Active , Mutation , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Receptors, Notch/metabolism
4.
J Cell Mol Med ; 26(3): 880-892, 2022 02.
Article in English | MEDLINE | ID: mdl-34931449

ABSTRACT

The aggregation of ß-amyloid peptide 42 results in the formation of toxic oligomers and plaques, which plays a pivotal role in Alzheimer's disease pathogenesis. Aß42 is one of several Aß peptides, all of Aß30 to Aß43 that are produced as a result of γ-secretase-mediated regulated intramembrane proteolysis of the amyloid precursor protein. γ-Secretase modulators (GSMs) represent a promising class of Aß42-lowering anti-amyloidogenic compounds for the treatment of AD. Gamma-secretase modulators change the relative proportion of secreted Aß peptides, while sparing the γ-secretase-mediated processing event resulting in the release of the cytoplasmic APP intracellular domain. In this study, we have characterized how GSMs affect the γ-secretase cleavage of three γ-secretase substrates, E-cadherin, ephrin type A receptor 4 (EphA4) and ephrin type B receptor 2 (EphB2), which all are implicated in important contexts of cell signalling. By using a reporter gene assay, we demonstrate that the γ-secretase-dependent generation of EphA4 and EphB2 intracellular domains is unaffected by GSMs. We also show that γ-secretase processing of EphA4 and EphB2 results in the release of several Aß-like peptides, but that only the production of Aß-like proteins from EphA4 is modulated by GSMs, but with an order of magnitude lower potency as compared to Aß modulation. Collectively, these results suggest that GSMs are selective for γ-secretase-mediated Aß production.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Humans , Mutation
5.
Neurosci Res ; 109: 9-15, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26907521

ABSTRACT

BACE1-mediated cleavage of APP is a pivotal step in the production of the Alzheimer related Aß peptide and inhibitors of BACE1 are currently in clinical development for the treatment of Alzheimer disease (AD). While processing and trafficking of APP has been extensively studied in non-neuronal cells, the fate of APP at neuronal synapses and in response to reduced BACE1 activity has not been fully elucidated. Here we examined the consequence of reduced BACE1 activity on endogenous synaptic APP by monitoring N- and C-terminal APP epitopes by immunocytochemistry. In control rodent primary hippocampal neuron cultures, labeling with antibodies directed to N-terminal APP epitopes showed a significant overlap with synaptic vesicle markers (SV2 or synaptotagmin). In contrast, labeling with antibodies directed to C-terminal epitopes of APP showed only a limited overlap with these proteins. In neurons derived from BACE1-deficient mice, and in control neurons treated with a BACE1 inhibitor, both the N-terminal and the C-terminal APP labeling overlapped significantly with synaptic vesicle markers. Moreover, BACE1 inhibition increased the proximity between the APP C-terminus and SV2 as shown by a proximity ligation assay. These results, together with biochemical observations, indicate that BACE1 can regulate the levels of full-length APP at neuronal synapses.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Neurons/drug effects , Synapses/drug effects , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Cells, Cultured , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Neurons/metabolism , Pyrimidines/pharmacology , Rats, Sprague-Dawley , Synapses/metabolism
6.
J Neurochem ; 132(4): 477-86, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25156639

ABSTRACT

Aggregation of amyloid beta (Aß) peptides and the subsequent neural plaque formation is a central aspect of Alzheimer's disease. Various strategies to reduce Aß load in the brain are therefore intensely pursued. It has been hypothesized that reducing Aß peptides in the periphery, that is in organs outside the brain, would be a way to diminish Aß levels and plaque load in the brain. In this report, we put this peripheral sink hypothesis to test by investigating how selective inhibition of Aß production in the periphery using a ß-secretase (BACE)1 inhibitor or reduced BACE1 gene dosage affects Aß load in the brain. Selective inhibition of peripheral BACE1 activity in wild-type mice or mice over-expressing amyloid precursor protein (APPswe transgenic mice; Tg2576) reduced Aß levels in the periphery but not in the brain, not even after chronic treatment over several months. In contrast, a BACE1 inhibitor with improved brain disposition reduced Aß levels in both brain and periphery already after acute dosing. Mice heterozygous for BACE1, displayed a 62% reduction in plasma Aß40, whereas brain Aß40 was only lowered by 11%. These data suggest that reduction of Aß in the periphery is not sufficient to reduce brain Aß levels and that BACE1 is not the rate-limiting enzyme for Aß processing in the brain. This provides evidence against the peripheral sink hypothesis and suggests that a decrease in Aß via BACE1 inhibition would need to be carried out in the brain. Aggregation of amyloid beta (Aß) peptides in the brain is a central aspect of Alzheimer's disease. In this study, we demonstrate that inhibition of Aß formation by BACE1 inhibitors needs to be carried out in the brain and that reduction of Aß in the periphery is not sufficient to reduce brain Aß levels. This information is useful for developing future Aß-targeting therapies for Alzheimer's disease.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Brain/enzymology , Animals , Brain/drug effects , Caco-2 Cells , Cricetinae , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
FEBS Open Bio ; 4: 393-406, 2014.
Article in English | MEDLINE | ID: mdl-24918054

ABSTRACT

The enzyme complex γ-secretase generates amyloid ß-peptide (Aß), a 37-43-residue peptide associated with Alzheimer disease (AD). Mutations in presenilin 1 (PS1), the catalytical subunit of γ-secretase, result in familial AD (FAD). A unifying theme among FAD mutations is an alteration in the ratio Aß species produced (the Aß42/Aß40 ratio), but the molecular mechanisms responsible remain elusive. In this report we have studied the impact of several different PS1 FAD mutations on the integration of selected PS1 transmembrane domains and on PS1 active site conformation, and whether any effects translate to a particular amyloid precursor protein (APP) processing phenotype. Most mutations studied caused an increase in the Aß42/Aß40 ratio, but via different mechanisms. The mutations that caused a particular large increase in the Aß42/Aß40 ratio did also display an impaired APP intracellular domain (AICD) formation and a lower total Aß production. Interestingly, seven mutations close to the catalytic site caused a severely impaired integration of proximal transmembrane/hydrophobic sequences into the membrane. This structural defect did not correlate to a particular APP processing phenotype. Six selected FAD mutations, all of which exhibited different APP processing profiles and impact on PS1 transmembrane domain integration, were found to display an altered active site conformation. Combined, our data suggest that FAD mutations affect the PS1 structure and active site differently, resulting in several complex APP processing phenotypes, where the most aggressive mutations in terms of increased Aß42/Aß40 ratio are associated with a decrease in total γ-secretase activity.

8.
Front Pharmacol ; 5: 102, 2014.
Article in English | MEDLINE | ID: mdl-24847271

ABSTRACT

Alzheimer's Disease (AD) is the most common form of dementia, affecting approximately 36 million people worldwide. To date there is no preventive or curative treatment available for AD, and in absence of major progress in therapeutic development, AD manifests a concrete socioeconomic threat. The awareness of the growing problem of AD is increasing, exemplified by the recent G8 Dementia Summit, a meeting held in order to set the stage and steer the compass for the future. Simultaneously, and paradoxically, we have seen key players in the pharmaceutical industry that have recently closed or significantly decreased their R&D spending on AD and other CNS disorders. Given the pressing need for new treatments in this area, other actors need to step-in and enter this drug discovery arena complementing the industrial efforts, in order to turn biological and technological progress into novel therapeutics. In this article, we present an example of a novel drug discovery initiative that in a non-profit setting, aims to integrate with both preclinical and clinical academic groups and pharmaceutical industry to explore the therapeutic potential of new concepts in patients, using novel biology, state of the art technologies and rapid concept testing.

9.
J Biol Chem ; 289(3): 1540-50, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24225948

ABSTRACT

Processing of the amyloid precursor protein (APP) by γ-secretase results in generation of Aß peptides of different lengths ranging from 51 to 30 residues. Accumulation of Aß and in particular Aß42 is enhanced by familial Alzheimer disease (FAD) causing mutations in APP and is believed to play a pivotal role. The molecular mechanism underlying normal Aß production, the impact of FAD mutations on this process and how anti-amyloidogenic γ-secretase modulators (GSMs) cause a selective decrease in Aß40 and Aß42 and an increase in shorter Aß peptides, however, is poorly understood. By using a combined immuno- and LC-MS-based assay we identify several major intermediates, i.e. 3- and 4-peptides that line up head to head across the entire APP transmembrane sequence from Aß51 to Aß31/Aß30 and from Aß49 to Aß30/31. FAD APP mutations displayed a relative increase in 3- and 4-peptides from Aß48 to Aß38 compared with Aß49 to Aß37. These findings correlate with an increase in the Aß42/40 ratio. GSMs caused a decrease in Aß40 and Aß42 and an increase in Aß37 and Aß38 paralleled by an increase of the intermediates Aß40-38 and Aß42-39. Collectively, these data provide a thorough characterization of all intermediate steps in Aß production in native cell membranes and provide key mechanistic insights to genetic and pharmacological modulation of Aß generation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Gene Expression Regulation , Genetic Diseases, Inborn/metabolism , Mutation , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , HEK293 Cells , Humans
10.
Eur J Clin Pharmacol ; 69(6): 1247-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23288352

ABSTRACT

PURPOSE: To date, γ-secretase inhibition is the most frequently studied mechanism of reducing Aß in clinical trials with as yet no therapeutic success for AD patients, as measured by the slowing down of cognitive decline or an improvement in cognitive function. The aims of this investigation were to evaluate whether the amyloid hypothesis has been tested clinically, and to explore whether preclinical data are predictive of clinical Aß effects. METHODS: A model-based-meta analysis on Aß levels and drug exposure over time was performed on published and in-house (pre-)clinical data with γ-secretase inhibitors (GSIs; semagacestat, avagacestat, begacestat, PF-3074014, and MK0752). RESULTS: The clinical data available did not show any significant or robust reduction of CNS Aß over time at dose levels intended for AD patients. In contrast, these doses resulted in an average increase in plasma Aß levels over a 24-h interval. A general agreement between preclinical and clinical data was found and allowed for interspecies extrapolations. CONCLUSIONS: More substantially, CNS Aß-lowering drugs are needed to test whether inhibition of Aß production is efficacious in mild AD. Predictions based on preclinical data could assist in the selection of drug candidates and trial design.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Brain/drug effects , Enzyme Inhibitors/therapeutic use , Models, Biological , Alzheimer Disease/enzymology , Alzheimer Disease/psychology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/blood , Animals , Brain/enzymology , Cognition/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Humans , Species Specificity , Treatment Outcome
11.
J Neurosci ; 32(48): 17297-305, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197721

ABSTRACT

γ-Secretase inhibition represents a major therapeutic strategy for lowering amyloid ß (Aß) peptide production in Alzheimer's disease (AD). Progress toward clinical use of γ-secretase inhibitors has, however, been hampered due to mechanism-based adverse events, primarily related to impairment of Notch signaling. The γ-secretase inhibitor MRK-560 represents an exception as it is largely tolerable in vivo despite displaying only a small selectivity between Aß production and Notch signaling in vitro. In exploring the molecular basis for the observed tolerability, we show that MRK-560 displays a strong preference for the presenilin 1 (PS1) over PS2 subclass of γ-secretases and is tolerable in wild-type mice but causes dose-dependent Notch-related side effect in PS2-deficient mice at drug exposure levels resulting in a substantial decrease in brain Aß levels. This demonstrates that PS2 plays an important role in mediating essential Notch signaling in several peripheral organs during pharmacological inhibition of PS1 and provide preclinical in vivo proof of concept for PS2-sparing inhibition as a novel, tolerable and efficacious γ-secretase targeting strategy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Brain/drug effects , Presenilin-2/metabolism , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Mice , Presenilin-2/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
12.
J Biol Chem ; 287(39): 32640-50, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22851182

ABSTRACT

The γ-secretase complex is an appealing drug target when the therapeutic strategy is to alter amyloid-ß peptide (Aß) aggregation in Alzheimer disease. γ-Secretase is directly involved in Aß formation and determines the pathogenic potential of Aß by generating the aggregation-prone Aß42 peptide. Because γ-secretase mediates cleavage of many substrates involved in cell signaling, such as the Notch receptor, it is crucial to sustain these pathways while altering the Aß secretion. A way of avoiding interference with the physiological function of γ-secretase is to use γ-secretase modulators (GSMs) instead of inhibitors of the enzyme. GSMs modify the Aß formation from producing the amyloid-prone Aß42 variant to shorter and less amyloidogenic Aß species. The modes of action of GSMs are not fully understood, and even though the pharmacology of GSMs has been thoroughly studied regarding Aß generation, knowledge is lacking about their effects on other substrates, such as Notch. Here, using immunoprecipitation followed by MALDI-TOF MS analysis, we found that two novel, second generation GSMs modulate both Notch ß and Aß production. Moreover, by correlating S3-specific Val-1744 cleavage of Notch intracellular domain (Notch intracellular domain) to total Notch intracellular domain levels using immunocytochemistry, we also demonstrated that Notch intracellular domain is not modulated by the compounds. Interestingly, two well characterized, nonsteroidal anti-inflammatory drugs (nonsteroidal anti-inflammatory drug), R-flurbiprofen and sulindac sulfide, affect only Aß and not Notch ß formation, indicating that second generation GSMs and nonsteroidal anti-inflammatory drug-based GSMs have different modes of action regarding Notch processing.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Flurbiprofen/pharmacology , Receptors, Notch/metabolism , Sulindac/analogs & derivatives , Amyloid/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Female , HEK293 Cells , Humans , Mice , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Protein Structure, Tertiary , Receptors, Notch/genetics , Sulindac/pharmacology
13.
PLoS One ; 7(2): e31084, 2012.
Article in English | MEDLINE | ID: mdl-22328928

ABSTRACT

BACE1 is a key enzyme for amyloid-ß (Aß) production, and an attractive therapeutic target in Alzheimer's disease (AD). Here we report that BACE1 inhibitors have distinct effects on neuronal Aß metabolism, inducing a unique pattern of secreted Aß peptides, analyzed in cell media from amyloid precursor protein (APP) transfected cells and in cerebrospinal fluid (CSF) from dogs by immunoprecipitation-mass spectrometry, using several different BACE1 inhibitors. Besides the expected reductions in Aß1-40 and Aß1-42, treatment also changed the relative levels of several other Aß isoforms. In particular Aß1-34 decreased, while Aß5-40 increased, and these changes were more sensitive to BACE1 inhibition than the changes in Aß1-40 and Aß1-42. The effects on Aß5-40 indicate the presence of a BACE1 independent pathway of APP degradation. The described CSF Aß pattern may be used as a pharmacodynamic fingerprint to detect biochemical effects of BACE1-therapies in clinical trials, which might accelerate development of novel therapies.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Central Nervous System/drug effects , Central Nervous System/metabolism , Cerebrospinal Fluid/metabolism , Enzyme Inhibitors/pharmacology , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line , Chromatography, Liquid , Dimethyl Sulfoxide/pharmacology , Dogs , Female , Humans , Immunoassay , Immunoprecipitation , Male , Mass Spectrometry , Tandem Mass Spectrometry
14.
J Biol Chem ; 287(15): 11810-9, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22334705

ABSTRACT

γ-Secretase-mediated cleavage of amyloid precursor protein (APP) results in the production of Alzheimer disease-related amyloid-ß (Aß) peptides. The Aß42 peptide in particular plays a pivotal role in Alzheimer disease pathogenesis and represents a major drug target. Several γ-secretase modulators (GSMs), such as the nonsteroidal anti-inflammatory drugs (R)-flurbiprofen and sulindac sulfide, have been suggested to modulate the Alzheimer-related Aß production by targeting the APP. Here, we describe novel GSMs that are selective for Aß modulation and do not impair processing of Notch, EphB2, or EphA4. The GSMs modulate Aß both in cell and cell-free systems as well as lower amyloidogenic Aß42 levels in the mouse brain. Both radioligand binding and cellular cross-competition experiments reveal a competitive relationship between the AstraZeneca (AZ) GSMs and the established second generation GSM, E2012, but a noncompetitive interaction between AZ GSMs and the first generation GSMs (R)-flurbiprofen and sulindac sulfide. The binding of a (3)H-labeled AZ GSM analog does not co-localize with APP but overlaps anatomically with a γ-secretase targeting inhibitor in rodent brains. Combined, these data provide compelling evidence of a growing class of in vivo active GSMs, which are selective for Aß modulation and have a different mechanism of action compared with the original class of GSMs described.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Azepines/pharmacology , Protein Processing, Post-Translational/drug effects , Pyrans/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Azepines/chemistry , Binding, Competitive , Brain/drug effects , Brain/metabolism , Carbamates/pharmacology , Cell-Free System , Dibenzazepines/pharmacology , Dipeptides/pharmacology , Drug Interactions , Female , Flurbiprofen/pharmacology , Guinea Pigs , HEK293 Cells , Humans , Imidazoles/pharmacology , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Protein Binding , Pyrans/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Rats , Receptor, EphA4/metabolism , Receptor, EphB2/metabolism , Receptors, Notch/metabolism , Sulfonamides/pharmacology , Sulindac/analogs & derivatives , Sulindac/pharmacology
16.
Biochem Biophys Res Commun ; 393(1): 21-7, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20097169

ABSTRACT

beta-Secretase (BACE) is an aspartyl protease, which proteolytically processes amyloid precursor protein, making BACE an interesting pharmacological target in Alzheimer's disease. To study the enzymatic function of BACE, we mutated either of the two aspartic acid residues in the active site of BACE. This rendered BACE functionally inactive without affecting the degree of glycosylation or endosomal localization. In contrast, substituting both active site aspartic acid residues produced a functionally inactive, endoplasmic reticulum-retained and partially glycosylated BACE. Interestingly, co-expression of the two single active site mutants partially restored beta-site cleavage of amyloid precursor protein, and the restored activity was inhibited with similar dose-dependency and potency as wildtype BACE by a small molecule inhibitor raised against BACE. In sum, our data suggest that two different active site mutants can complement each other in a partially functional BACE dimer and mediate APP processing.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Catalytic Domain/genetics , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Knockout , Mutation , Protein Multimerization
17.
J Biol Chem ; 285(12): 8527-36, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20106965

ABSTRACT

Gamma-secretase is an enzyme complex that mediates both Notch signaling and beta-amyloid precursor protein (APP) processing, resulting in the generation of Notch intracellular domain, APP intracellular domain, and the amyloid beta peptide (Abeta), the latter playing a central role in Alzheimer disease (AD). By a hitherto undefined mechanism, the activity of gamma-secretase gives rise to Abeta peptides of different lengths, where Abeta42 is considered to play a particular role in AD. In this study we have examined the role of the large hydrophilic loop (amino acids 320-374, encoded by exon 10) of presenilin 1 (PS1), the catalytic subunit of gamma-secretase, for gamma-secretase complex formation and activity on Notch and APP processing. Deletion of exon 10 resulted in impaired PS1 endoproteolysis, gamma-secretase complex formation, and had a differential effect on Abeta-peptide production. Although the production of Abeta38, Abeta39, and Abeta40 was severely impaired, the effect on Abeta42 was affected to a lesser extent, implying that the production of the AD-related Abeta42 peptide is separate from the production of the Abeta38, Abeta39, and Abeta40 peptides. Interestingly, formation of the intracellular domains of both APP and Notch was intact, implying a differential cleavage activity between the epsilon/S3 and gamma sites. The most C-terminal amino acids of the hydrophilic loop were important for regulating APP processing. In summary, the large hydrophilic loop of PS1 appears to differentially regulate the relative production of different Abeta peptides without affecting Notch processing, two parameters of significance when considering gamma-secretase as a target for pharmaceutical intervention in AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/chemistry , Presenilin-1/chemistry , Presenilin-1/physiology , Receptors, Notch/metabolism , Blastocyst/metabolism , Catalytic Domain , Cell Line , DNA, Complementary/metabolism , Exons , Glycoside Hydrolases/metabolism , Humans , Peptide Fragments/chemistry , Protein Conformation , Protein Structure, Tertiary , Transfection
18.
Curr Opin Pharmacol ; 7(1): 112-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17169612

ABSTRACT

Data accumulated during the past two decades place amyloid beta-peptide (Abeta) at center stage as the main perpetrator in initiating the pathological cascade that eventually leads to Alzheimer's disease. Consequently, significant resources have been allocated to identify and develop treatment strategies that alter the metabolism of Abeta. The gamma-secretase protease has deservedly received attention as an attractive drug target, as it is directly involved in Abeta biogenesis and determines the pathogenic potential of Abeta by its heterogeneous catalytic action, generating peptides of various lengths. Despite the complexity of the multi-subunit gamma-secretase and the lack of structural information, drug discovery research has identified small-molecule compounds that inhibit or modulate activity of this enzyme and some of these have already entered clinical trials.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Humans
19.
Dev Cell ; 9(5): 617-28, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16256737

ABSTRACT

In addition to controlling a switch to glycolytic metabolism and induction of erythropoiesis and angiogenesis, hypoxia promotes the undifferentiated cell state in various stem and precursor cell populations. Here, we show that the latter process requires Notch signaling. Hypoxia blocks neuronal and myogenic differentiation in a Notch-dependent manner. Hypoxia activates Notch-responsive promoters and increases expression of Notch direct downstream genes. The Notch intracellular domain interacts with HIF-1alpha, a global regulator of oxygen homeostasis, and HIF-1alpha is recruited to Notch-responsive promoters upon Notch activation under hypoxic conditions. Taken together, these data provide molecular insights into how reduced oxygen levels control the cellular differentiation status and demonstrate a role for Notch in this process.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carbamates/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Dipeptides/pharmacology , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/drug effects , Receptors, Notch/genetics , Repressor Proteins/drug effects , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Transcription Factor HES-1
20.
J Neurochem ; 95(3): 880-90, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16135086

ABSTRACT

The gamma-secretase complex processes substrate proteins within membranes and consists of four proteins: presenilin (PS), nicastrin, Aph-1 and Pen-2. PS harbours the enzymatic activity of the complex, and there are two mammalian PS homologues: PS1 and PS2. PS undergoes endoproteolysis, generating the N- and C-terminal fragments, NTF and CTF, which represent the active species of PS. To characterize the functional similarity between complexes of various PS composition, we analysed PS1, PS2, and chimeric PS composed of the NTF from PS1 and CTF from PS2, or vice versa, in assembly and function of the gamma-secretase complex. Chimeric PSs, like PS1 and PS2, undergo normal endoproteolysis when introduced into cells devoid of endogenous PS. Furthermore, PS2 CTF can, at least partially, restore processing in a truncated PS1, which cannot undergo endoproteolysis. All PS forms enable maturation of nicastrin and cleave full length Notch receptors, indicating that both PS1 and PS2 are present at the cell surface. Finally, when co-introduced as separate molecules, NTF and CTF of different PS origin reconstitute gamma-secretase activity. In conclusion, these data show that endoproteolysis, NTF-CTF interactions, and the assembly and activity of gamma-secretase complexes are very conserved between PS1 and PS2.


Subject(s)
Endopeptidases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Cell Line , Endopeptidases/chemistry , Endopeptidases/metabolism , Enzyme Activation/physiology , Gene Expression Regulation, Enzymologic , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Presenilin-1 , Presenilin-2 , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...