Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Cell Rep ; 13(4): 812-828, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26489457

ABSTRACT

Axon guidance relies on precise translation of extracellular signal gradients into local changes in cytoskeletal dynamics, but the molecular mechanisms regulating dose-dependent responses of growth cones are still poorly understood. Here, we show that during embryonic development in growing axons, a low level of Semaphorin3A stimulation is buffered by the prolyl isomerase Pin1. We demonstrate that Pin1 stabilizes CDK5-phosphorylated CRMP2A, the major isoform of CRMP2 in distal axons. Consequently, Pin1 knockdown or knockout reduces CRMP2A levels specifically in distal axons and inhibits axon growth, which can be fully rescued by Pin1 or CRMP2A expression. Moreover, Pin1 knockdown or knockout increases sensitivity to Sema3A-induced growth cone collapse in vitro and in vivo, leading to developmental abnormalities in axon guidance. These results identify an important isoform-specific function and regulation of CRMP2A in controlling axon growth and uncover Pin1-catalyzed prolyl isomerization as a regulatory mechanism in axon guidance.


Subject(s)
Axons/metabolism , Nerve Tissue Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Immunoprecipitation , Male , NIMA-Interacting Peptidylprolyl Isomerase , Nerve Tissue Proteins/genetics , Peptidylprolyl Isomerase/genetics , Phosphorylation , Signal Transduction , Zebrafish , Zebrafish Proteins/genetics
2.
Neuron ; 75(5): 824-37, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22958823

ABSTRACT

Although Aß peptides are causative agents in Alzheimer's disease (AD), the underlying mechanisms are still elusive. We report that Aß42 induces a translational block by activating AMPK, thereby inhibiting the mTOR pathway. This translational block leads to widespread ER stress, which activates JNK3. JNK3 in turn phosphorylates APP at T668, thereby facilitating its endocytosis and subsequent processing. In support, pharmacologically blocking translation results in a significant increase in Aß42 in a JNK3-dependent manner. Thus, JNK3 activation, which is increased in human AD cases and a familial AD (FAD) mouse model, is integral to perpetuating Aß42 production. Concomitantly, deletion of JNK3 from FAD mice results in a dramatic reduction in Aß42 levels and overall plaque loads and increased neuronal number and improved cognition. This reveals AD as a metabolic disease that is under tight control by JNK3.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Peptide Fragments/metabolism , Stress, Physiological/physiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/toxicity , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred Strains , Mice, Knockout , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , Organ Culture Techniques , Peptide Fragments/biosynthesis , Peptide Fragments/toxicity , Primary Cell Culture , Rats
3.
J Alzheimers Dis ; 30(2): 277-97, 2012.
Article in English | MEDLINE | ID: mdl-22430533

ABSTRACT

Increased amyloidogenic processing of the amyloid-ß protein precursor (AßPP) is a characteristic of Alzheimer's disease (AD). We previously observed that the prolyl isomerase Pin1, which is down-regulated in AD, regulates AßPP conformation accelerating cis/trans isomerization of the phospho-Thr668-Pro669 peptide bond, and that Pin1 knockout in mice increases the amyloidogenic processing of AßPP, although the underlying mechanism is still unknown. Since the intracellular localization of AßPP determines whether the processing will be amyloidogenic or non-amyloidogenic, here we addressed the question whether loss of Pin1 function affects the intracellular localization of AßPP, influencing AßPP processing. Using cellular models of Pin1 knockout and Pin1 knockdown, we have demonstrated that lowering Pin1 levels changed the intracellular localization and the processing of AßPP. Under these conditions, less AßPP was retained at the plasma membrane favoring the amyloidogenic processing, and the kinetics of AßPP internalization increased as well as the nuclear trafficking of AßPP C-terminal fragment AICD. In addition, AßPPThr668Ala mutant, which cannot bind to Pin1 and retains more trans conformation, rescued the levels of AßPP at the plasma membrane in Pin1 knockout cells. Thus, loss of Pin1 function contributes to amyloidogenic pathways, by facilitating both the removal of AßPP from compartments where it is mostly non-amyloidogenic and its internalization to more amyloidogenic compartments. These data suggest that physiological levels of Pin1 are important to control the intracellular localization and metabolic fate of Thr668-phosphorylated AßPP, and regulation of AßPP conformation is especially important in pathologic conditions of AßPP hyperphosphorylation and/or loss of Pin1 function, associated with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloidosis/metabolism , Peptidylprolyl Isomerase/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloidosis/pathology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cricetinae , Endocytosis/physiology , Female , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Phosphorylation/physiology , Protein Structure, Tertiary , Protein Transport/physiology
4.
Neurobiol Aging ; 33(4): 804-13, 2012 Apr.
Article in English | MEDLINE | ID: mdl-20580132

ABSTRACT

Alzheimer's disease (AD), the most common form of dementia, is characterized by the presence of neurofibrillary tangles composed of tau and senile plaques of amyloid-beta peptides (Aß) derived from amyloid precursor protein (APP). Pin1 is a unique prolyl isomerase that has been shown to protect against age-dependent neurodegeneration by acting on phosphorylated tau and APP to suppress tangle formation and amyloidogenic APP processing. Here we report a functional polymorphism, rs2287839, in the Pin1 promoter that is significantly associated with a 3-year delay in the average age at onset (AAO) of late-onset AD in a Chinese population. More significantly, the Pin1 polymorphism rs2287839 is located within the consensus binding motif for the brain-selective transcription factor, AP4 (CAGCTG) and almost completely abolishes the ability of AP4 to bind and suppress the Pin1 promoter, as shown by chromatin immunoprecipitation, electrophoretic mobility shift assay, and promoter luciferase assay. Moreover, overexpression or knockdown of AP4 resulted in an 80% reduction or 2-fold increase in endogenous Pin1 levels, respectively. Thus, AP4 is a novel transcriptional repressor of Pin1 expression and the Pin1 promoter single nucleotide polymorphism (SNP) identified in this study that prevents such suppression is associated with delayed onset of AD. These results indicate that regulation of Pin1 by AP4 plays a critical role in determining age at onset of AD and might be a novel therapeutic target to delay the onset of AD.


Subject(s)
Alzheimer Disease/genetics , Peptidylprolyl Isomerase/genetics , Polymorphism, Single Nucleotide/genetics , Aged , Aged, 80 and over , Asian People/genetics , Cell Line, Transformed , Chi-Square Distribution , Chromatin Immunoprecipitation , Computational Biology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Electrophoretic Mobility Shift Assay , Female , Gene Expression Regulation/genetics , Gene Knockout Techniques , Genotype , Hong Kong , Humans , Male , Mental Status Schedule , NIMA-Interacting Peptidylprolyl Isomerase , Neurons/drug effects , Neurons/metabolism , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Regression Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology , Transfection
5.
J Biol Chem ; 287(10): 6969-73, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22184106

ABSTRACT

Alzheimer disease (AD) is characterized by the presence of senile plaques of amyloid-ß (Aß) peptides derived from amyloid precursor protein (APP) and neurofibrillary tangles made of hyperphosphorylated Tau. Increasing APP gene dosage or expression has been shown to cause familial early-onset AD. However, whether and how protein stability of APP is regulated is unclear. The prolyl isomerase Pin1 and glycogen synthase kinase-3ß (GSK3ß) have been shown to have the opposite effects on APP processing and Tau hyperphosphorylation, relevant to the pathogenesis of AD. However, nothing is known about their relationship. In this study, we found that Pin1 binds to the pT330-P motif in GSK3ß to inhibit its kinase activity. Furthermore, Pin1 promotes protein turnover of APP by inhibiting GSK3ß activity. A point mutation either at Thr-330, the Pin1-binding site in GSK3ß, or at Thr-668, the GSK3ß phosphorylation site in APP, abolished the regulation of GSK3ß activity, Thr-668 phosphorylation, and APP stability by Pin1, resulting in reduced non-amyloidogenic APP processing and increased APP levels. These results uncover a novel role of Pin1 in inhibiting GSK3ß kinase activity to reduce APP protein levels, providing a previously unrecognized mechanism by which Pin1 protects against Alzheimer disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Glycogen Synthase Kinase 3/metabolism , Peptidylprolyl Isomerase/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/genetics , Cell Line , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Phosphorylation/genetics , Point Mutation , Protein Stability , tau Proteins/genetics , tau Proteins/metabolism
6.
Expert Rev Mol Med ; 13: e21, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21682951

ABSTRACT

Phosphorylation of proteins on serine or threonine residues preceding proline is a key signalling mechanism in diverse physiological and pathological processes. Pin1 (peptidyl-prolyl cis-trans isomerase) is the only enzyme known that can isomerise specific Ser/Thr-Pro peptide bonds after phosphorylation and regulate their conformational changes with high efficiency. These Pin1-catalysed conformational changes can have profound effects on phosphorylation signalling by regulating a spectrum of target activities. Interestingly, Pin1 deregulation is implicated in a number of diseases, notably ageing and age-related diseases, including cancer and Alzheimer disease. Pin1 is overexpressed in most human cancers; it activates numerous oncogenes or growth enhancers and also inactivates a large number of tumour suppressors or growth inhibitors. By contrast, ablation of Pin1 prevents cancer, but eventually leads to premature ageing and neurodegeneration. Consistent with its neuroprotective role, Pin1 has been shown to be inactivated in neurons of patients with Alzheimer disease. Therefore, Pin1-mediated phosphorylation-dependent prolyl isomerisation represents a unique signalling mechanism that has a pivotal role in the development of human diseases, and might offer an attractive new diagnostic and therapeutic target.


Subject(s)
Aging/metabolism , Alzheimer Disease/enzymology , Neoplasms/enzymology , Peptidylprolyl Isomerase/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , Oxidative Stress , Peptidylprolyl Isomerase/chemistry , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Telomere
7.
Nat Cell Biol ; 11(1): 97-105, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19060891

ABSTRACT

Telomeres are essential for maintaining cellular proliferative capacity and their loss has been implicated in ageing. A key regulator in telomere maintenance is the telomeric protein TRF1, which was also identified as Pin2 in a screen for Pin1. Pin1 is a unique prolyl isomerase that regulates protein conformation and function after phosphorylation. However, little is known about the role of Pin1 in telomere regulation or the modulation of TRF1 by upstream signals. Here we identify TRF1 as a major conserved substrate for Pin1 during telomere maintenance and ageing. Pin1 inhibition renders TRF1 resistant to protein degradation, enhances TRF1 binding to telomeres, and leads to gradual telomere loss in human cells and in mice. Pin1-deficient mice also show widespread premature ageing phenotypes within just one generation, similar to those in telomerase-deficient mice after 4-5 consecutive generations. Thus, Pin1 is an essential regulator of TRF1 stability, telomere maintenance and ageing.


Subject(s)
Aging , Cellular Senescence , Peptidylprolyl Isomerase/physiology , Telomere/genetics , Telomeric Repeat Binding Protein 1/metabolism , Aging/genetics , Aging/metabolism , Animals , Cell Line , Cell Line, Tumor , Cellular Senescence/genetics , Chromosomal Instability/genetics , Humans , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Protein Binding/genetics , Signal Transduction/genetics , Telomerase/deficiency , Telomerase/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 1/genetics
8.
J Clin Invest ; 118(5): 1877-89, 2008 May.
Article in English | MEDLINE | ID: mdl-18431510

ABSTRACT

Tau pathology is a hallmark of many neurodegenerative diseases including Alzheimer disease (AD) and frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17). Genetic tau mutations can cause FTDP-17, and mice overexpressing tau mutants such as P301L tau are used as AD models. However, since no tau mutations are found in AD, it remains unclear how appropriate tau mutant mice are as an AD model. The prolyl isomerase Pin1 binds and isomerizes tau and has been implicated in protecting against neurodegeneration, but whether such Pin1 regulation is affected by tau mutations is unknown. Consistent with earlier findings that Pin1 KO induces tauopathy, here we demonstrate that Pin1 knockdown or KO increased WT tau protein stability in vitro and in mice and that Pin1 overexpression suppressed the tauopathy phenotype in WT tau transgenic mice. Unexpectedly, Pin1 knockdown or KO decreased P301L tau protein stability and abolished its robust tauopathy phenotype in mice. In contrast, Pin1 overexpression exacerbated the tauopathy phenotype in P301L tau mice. Thus, Pin1 has opposite effects on the tauopathy phenotype depending on whether the tau is WT or a P301L mutant, indicating the need for disease-specific therapies for tauopathies.


Subject(s)
Peptidylprolyl Isomerase/metabolism , Point Mutation , Tauopathies , tau Proteins , Animals , Brain/cytology , Brain/metabolism , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NIMA-Interacting Peptidylprolyl Isomerase , Neurons/cytology , Neurons/metabolism , Peptidylprolyl Isomerase/genetics , Phenotype , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/pathology , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/genetics , tau Proteins/metabolism
9.
Biochim Biophys Acta ; 1772(4): 422-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17317113

ABSTRACT

Presence of neuritic plaques and neurofibrillary tangles in the brain are two neuropathological hallmarks of Alzheimer's disease (AD), although the molecular basis of their coexistence remains elusive. The neurofibrillary tangles are composed of microtubule binding protein Tau, whereas neuritic plaques consist of amyloid-beta peptides derived from amyloid precursor protein (APP). Recently, the peptidyl-prolyl cis/trans isomerase Pin1 has been identified to regulate the function of certain proteins after phosphorylation and to play an important role in cell cycle regulation and cancer development. New data indicate that Pin1 also regulates the function and processing of Tau and APP, respectively, and is important for protecting against age-dependent neurodegeneration. Furthermore, Pin1 is the only gene known so far that, when deleted in mice, can cause both Tau and Abeta-related pathologies in an age-dependent manner, resembling many aspects of human Alzheimer's disease. Moreover, in the human AD brain Pin1 is downregulated or inhibited by oxidative modifications and/or genetic changes. These results suggest that Pin1 deregulation may provide a link between formation of tangles and plaques in AD.


Subject(s)
Alzheimer Disease/enzymology , Peptidylprolyl Isomerase/metabolism , Brain/enzymology , Humans , Models, Neurological , NIMA-Interacting Peptidylprolyl Isomerase , Neurons/enzymology , Reference Values , Tauopathies/enzymology
10.
Eur J Pharmacol ; 545(1): 29-38, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16904664

ABSTRACT

Alzheimer's disease is a progressive neurodegenerative disorder associated with aging and characterized by neurofibrillary tangles and amyloid plaques that deposit in the brain, triggering the neurodegenerative phenomena and leading to neuronal death. Amyloid plaques are primarily composed of beta-amyloid peptides, which derive from the Amyloid Precursor Protein (APP) upon the consequential action of beta- and gamma-secretase. This review discusses recent literature on beta- and gamma-secretase, and on those cellular factors, like cholesterol and phosphorylation of APP, that are involved in aging and may affect the function of both beta- and gamma-secretase.


Subject(s)
Alzheimer Disease/metabolism , Aging/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cholesterol/metabolism , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Phosphorylation , Presenilins/metabolism
11.
Nature ; 440(7083): 528-34, 2006 Mar 23.
Article in English | MEDLINE | ID: mdl-16554819

ABSTRACT

Neuropathological hallmarks of Alzheimer's disease are neurofibrillary tangles composed of tau and neuritic plaques comprising amyloid-beta peptides (Abeta) derived from amyloid precursor protein (APP), but their exact relationship remains elusive. Phosphorylation of tau and APP on certain serine or threonine residues preceding proline affects tangle formation and Abeta production in vitro. Phosphorylated Ser/Thr-Pro motifs in peptides can exist in cis or trans conformations, the conversion of which is catalysed by the Pin1 prolyl isomerase. Pin1 has been proposed to regulate protein function by accelerating conformational changes, but such activity has never been visualized and the biological and pathological significance of Pin1 substrate conformations is unknown. Notably, Pin1 is downregulated and/or inhibited by oxidation in Alzheimer's disease neurons, Pin1 knockout causes tauopathy and neurodegeneration, and Pin1 promoter polymorphisms appear to associate with reduced Pin1 levels and increased risk for late-onset Alzheimer's disease. However, the role of Pin1 in APP processing and Abeta production is unknown. Here we show that Pin1 has profound effects on APP processing and Abeta production. We find that Pin1 binds to the phosphorylated Thr 668-Pro motif in APP and accelerates its isomerization by over 1,000-fold, regulating the APP intracellular domain between two conformations, as visualized by NMR. Whereas Pin1 overexpression reduces Abeta secretion from cell cultures, knockout of Pin1 increases its secretion. Pin1 knockout alone or in combination with overexpression of mutant APP in mice increases amyloidogenic APP processing and selectively elevates insoluble Abeta42 (a major toxic species) in brains in an age-dependent manner, with Abeta42 being prominently localized to multivesicular bodies of neurons, as shown in Alzheimer's disease before plaque pathology. Thus, Pin1-catalysed prolyl isomerization is a novel mechanism to regulate APP processing and Abeta production, and its deregulation may link both tangle and plaque pathologies. These findings provide new insight into the pathogenesis and treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Processing, Post-Translational , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , CHO Cells , Catalysis , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Threonine/metabolism , Transfection
12.
Exp Neurol ; 179(2): 150-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12618121

ABSTRACT

BACE1 and BACE2 are recently discovered enzymes participating in processing of amyloid beta precursor protein (AbetaPP). Their discovery is contributing importantly to understanding the mechanism of amyloid-beta generation, and hence the pathogenesis of Alzheimer's disease (AD). Sporadic inclusion-body myositis (s-IBM) and hereditary inclusion-body myopathy (h-IBM) are progressive muscle diseases in which overproduction of AbetaPP and accumulation of its presumably toxic proteolytic product amyloid-beta (Abeta) in abnormal muscle fibers appear to play an important upstream role in the pathogenic cascade. In normal human muscle AbetaPP was also shown to be present and presumably playing a role (a) at neuromuscular junctions and (b) during muscle development. To investigate whether BACE1 and BACE2 play a role in normal and diseased human muscle, we have now studied them by immunocytochemistry and immunoblotting in 35 human muscle biopsies, including: 5 s-IBM; 5 chromosome-9p1-linked quadriceps-sparing h-IBM; and 25 control muscle biopsies. In addition, expression of BACE1 and BACE2 was studied in normal cultured human muscle. Our studies demonstrate that BACE1 and BACE2 (a) are expressed in normal adult muscle at the postsynaptic domain of neuromuscular junctions, and in cultured human muscle; (b) are accumulated in the form of plaque-like inclusions in both s-IBM and h-IBM vacuolated muscle fibers; and (c) are immunoreactive in necrotizing muscle fibers. Accordingly, BACE1 and BACE2 participate in normal and abnormal processes of human muscle, suggesting that their functions are broader than previously thought.


Subject(s)
Aspartic Acid Endopeptidases/biosynthesis , Muscle, Skeletal/enzymology , Myositis, Inclusion Body/enzymology , Myositis/enzymology , Amyloid Precursor Protein Secretases , Biopsy , Cells, Cultured , Endopeptidases , Humans , Immunoblotting , Immunohistochemistry , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/pathology , Myositis/genetics , Myositis/pathology , Myositis, Inclusion Body/pathology
13.
Mol Med ; 8(2): 67-74, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12080182

ABSTRACT

BACKGROUND: Members of membrane-bound disintegrin metalloproteinases (ADAMs) were shown to be capable of cleaving amyloid precursor protein (APP) at the alpha-cleavage site in different cell systems. One of the candidate alpha-secretases identified in this family is ADAM10. The present study addresses the following major questions: 1) Are the levels of an alpha-secretase candidate (i.e., ADAM10) reduced in accessible cells of Alzheimer Disease (AD) patients? 2) Are ADAM10 levels in the peripheral cells of AD patients related to a concomitant decrease in alpha APPs? MATERIALS AND METHODS: Western Blot analysis of ADAM10 is performed on platelet homogenates from 33 sporadic AD patients and on 26 age-matched control subjects. Moreover, the levels of alpha-secretase metabolite (alpha APPs) are tested both in platelets and cerebrospinal fluid (CSF) of the same pool of subjects by means of Western blot with a specific antibody. RESULTS: A significant decrease of platelet ADAM10 levels is observed in patients affected by probable AD when compared to control subjects and this is paralleled by a reduced level of alpha APPs released from platelets. Moreover, in the same pool of AD patients, alpha APPs levels were reduced concomitantly in CSF. CONCLUSIONS: ADAM10 is expressed in platelets. A reduced level of ADAM10 is observed in platelets obtained from AD patients compared to age-matched controls. Further, in the same pool of AD patients, a qualitatively and quantitatively similar decrease in alpha APPs is present both in thrombin-activated platelets and CSF, thus suggesting that alterations of APP processing might occur both in the neuronal compartment and peripheral cells.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Protein Precursor/blood , Amyloid beta-Protein Precursor/cerebrospinal fluid , Membrane Proteins/blood , Membrane Proteins/cerebrospinal fluid , Metalloendopeptidases/blood , Metalloendopeptidases/cerebrospinal fluid , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , ADAM Proteins , ADAM10 Protein , Aged , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/analysis , Blood Platelets/chemistry , Blotting, Western , Cerebral Cortex/chemistry , Electrophoresis, Polyacrylamide Gel , Female , Glycosylation , Humans , Male , Matched-Pair Analysis , Membrane Proteins/analysis , Metalloendopeptidases/analysis , Middle Aged , Peptide Fragments/analysis , Platelet Activation , Precipitin Tests
14.
Eur Neuropsychopharmacol ; 12(3): 195-200, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12007670

ABSTRACT

Three major amyloid precursor protein (APP) forms with apparent molecular weight ranging from 106 to 130 kDa are present in human platelets. Alzheimer disease (AD) is associated with a decreased APP forms ratio (APPr) between the three major forms. A total of 25 mild to moderate AD patients were investigated. Platelet APPr was studied before and after 30 days of acetylcholinesterase-inhibitor treatment (donepezil, 5 mg daily). Patients were grouped into non-epsilon4 carriers and epsilon4 carriers according to apolipoprotein E (ApoE) genotype. At baseline, all patients showed low APPr levels and no significant difference was found between the two ApoE subgroups. After treatment, although a marked improvement in APPr was observed in most patients, non-epsilon4 carriers displayed a higher increase compared to epsilon4 carriers (P<0.0001). The present study provides evidence that donepezil influences APP metabolism in platelets, and suggests that ApoE genotype might be an important modulating factor for drug responsiveness in AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/genetics , Cholinesterase Inhibitors/therapeutic use , Indans/therapeutic use , Piperidines/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/blood , Amyloid beta-Protein Precursor/blood , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Blood Platelets/metabolism , Donepezil , Female , Genotype , Humans , Longitudinal Studies , Male , Nootropic Agents/therapeutic use , Regression Analysis
15.
Mol Cell Neurosci ; 19(2): 175-85, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11860271

ABSTRACT

BACE (beta-site APP cleaving enzyme) has been recently proposed as the major aspartyl protease displaying beta secretase activity in neurons. The C-terminal domain of BACE contains a dileucine motif (LL499/500) that can potentially regulate its trafficking and endocytosis, and an adjacent serine, which is a potential phosphorylation site (S498) that could modulate the activity of the LL motif. In this paper we show that S498 is phosphorylated by casein kinase 1 (CKI). Mutating the LL to dialanine (AA) caused an increase in the levels of mature BACE. The LL to AA mutation increased levels of BACE on the cell surface and decreased the internalization of BACE. Mutating the S498 to alanine did not alter levels of cell surface BACE. Mutating either the leucines or the serine did not alter the secretion of A(beta). Our data are consistent with a role for the cytoplasmic domain in regulating BACE trafficking and localization.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Mutation/physiology , Protein Transport/physiology , Signal Transduction/physiology , Amino Acid Sequence/physiology , Amyloid Precursor Protein Secretases , Animals , Brain/enzymology , COS Cells , Dimerization , Endopeptidases , Leucine/metabolism , Phosphorylation , Protein Structure, Tertiary/physiology , Serine/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL