251
|
Binnington JC, Kalisch BE. Nitric oxide synthase inhibitors modulate nerve growth factor-mediated regulation of amyloid precursor protein expression in PC12 cells. J Neurochem 2007; 101:422-33. [PMID: 17402971 DOI: 10.1111/j.1471-4159.2006.04378.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Nerve growth factor (NGF) can regulate nitric oxide synthase (NOS) expression and nitric oxide (NO) can modulate NGF-mediated neurotrophic responses. In this study, the role of NO in NGF-stimulated amyloid precursor protein (APP) levels was studied. PC12 cells were treated with either the non-selective NOS inhibitor N(omega)-nitro-L-arginine methylester (L-NAME) or the inducible NOS selective inhibitor s-methylisothiourea (S-MIU), and the effect on NGF-mediated increases in APP expression was determined. NGF significantly increased total APP protein levels following 96 h of treatment and this increase was prevented in cells pre-treated with S-MIU. Pre-treatment of cells with actinomycin D also blocked this NGF-mediated induction of APP, indicating de novo protein synthesis is necessary. Treatment with NGF increased APP promoter activity; however, this increase was only partially inhibited by pre-treatment with S-MIU and was increased in the presence of L-NAME. This suggests that NO may be modulating other aspects of APP expression in addition to transcription. Inhibition of NGF signaling pathways was also investigated using inhibitors of mitogen-activated protein (MAP) kinase (U0126), Akt (LY294002) and protein kinase C (PKC; U73122 and bisindolylmaleimide 1 (BIS-1)) activation. Inhibition of each of these pathways prevented NGF-mediated increases in APP protein expression; however, only BIS-1 attenuated NGF-mediated increases in promoter activation. This study indicates that NO is involved in the NGF-mediated regulation of APP, in part at the level of APP transcription and could involve the modulation of NGF signal transduction pathways.
Collapse
Affiliation(s)
- Janet C Binnington
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
252
|
Kong GKW, Adams JJ, Harris HH, Boas JF, Curtain CC, Galatis D, Masters CL, Barnham KJ, McKinstry WJ, Cappai R, Parker MW. Structural Studies of the Alzheimer’s Amyloid Precursor Protein Copper-binding Domain Reveal How it Binds Copper Ions. J Mol Biol 2007; 367:148-61. [PMID: 17239395 DOI: 10.1016/j.jmb.2006.12.041] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Revised: 12/11/2006] [Accepted: 12/15/2006] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the major cause of dementia. Amyloid beta peptide (Abeta), generated by proteolytic cleavage of the amyloid precursor protein (APP), is central to AD pathogenesis. APP can function as a metalloprotein and modulate copper (Cu) transport, presumably via its extracellular Cu-binding domain (CuBD). Cu binding to the CuBD reduces Abeta levels, suggesting that a Cu mimetic may have therapeutic potential. We describe here the atomic structures of apo CuBD from three crystal forms and found they have identical Cu-binding sites despite the different crystal lattices. The structure of Cu(2+)-bound CuBD reveals that the metal ligands are His147, His151, Tyr168 and two water molecules, which are arranged in a square pyramidal geometry. The site resembles a Type 2 non-blue Cu center and is supported by electron paramagnetic resonance and extended X-ray absorption fine structure studies. A previous study suggested that Met170 might be a ligand but we suggest that this residue plays a critical role as an electron donor in CuBDs ability to reduce Cu ions. The structure of Cu(+)-bound CuBD is almost identical to the Cu(2+)-bound structure except for the loss of one of the water ligands. The geometry of the site is unfavorable for Cu(+), thus providing a mechanism by which CuBD could readily transfer Cu ions to other proteins.
Collapse
Affiliation(s)
- Geoffrey K-W Kong
- Biota Structural Biology Laboratory, St. Vincent's Institute, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
253
|
Zinser EG, Hartmann T, Grimm MOW. Amyloid beta-protein and lipid metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:1991-2001. [PMID: 17418089 DOI: 10.1016/j.bbamem.2007.02.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 02/14/2007] [Accepted: 02/15/2007] [Indexed: 01/16/2023]
Abstract
Lipids play an important part as risk or protective factors for Alzheimer's disease. This review summarizes the current findings in which lipids influence Alzheimer's disease and introduces the molecular mechanism how these lipids are linked to amyloid production. Besides the pathological impact of amyloid in Alzheimer's disease, amyloid has a physiological function in regulating lipid homeostasis in return. The understanding of the resulting regulatory cycles between amyloid precursor protein processing and lipids provides a platform for the development of new causal therapeutic approaches for Alzheimer's disease.
Collapse
Affiliation(s)
- Eva G Zinser
- Universität des Saarlandes, Uniklinikum Homburg, Neurobiologie, Neurologie, Gebäude 90, 66421 Homburg/Saar, Germany
| | | | | |
Collapse
|
254
|
Gralle M, Ferreira ST. Structure and functions of the human amyloid precursor protein: the whole is more than the sum of its parts. Prog Neurobiol 2007; 82:11-32. [PMID: 17428603 DOI: 10.1016/j.pneurobio.2007.02.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 10/26/2006] [Accepted: 02/01/2007] [Indexed: 12/30/2022]
Abstract
The amyloid precursor protein (APP) is a transmembrane protein that plays major roles in the regulation of several important cellular functions, especially in the nervous system, where it is involved in synaptogenesis and synaptic plasticity. The secreted extracellular domain of APP, sAPPalpha, acts as a growth factor for many types of cells and promotes neuritogenesis in post-mitotic neurons. Alternative proteolytic processing of APP releases potentially neurotoxic species, including the amyloid-beta (Abeta) peptide that is centrally implicated in the pathogenesis of Alzheimer's disease (AD). Reinforcing this biochemical link to neuronal dysfunction and neurodegeneration, APP is also genetically linked to AD. In this review, we discuss the biological functions of APP in the context of tissue morphogenesis and restructuring, where APP appears to play significant roles both as a contact receptor and as a diffusible factor. Structural investigation of APP, which is necessary for a deeper understanding of its roles at a molecular level, has also been advancing rapidly. We summarize recent progress in the determination of the structure of isolated APP fragments and of the conformations of full-length sAPPalpha, in both monomeric and dimeric states. The potential role of APP dimerization for the regulation of its biological functions is also discussed.
Collapse
Affiliation(s)
- Matthias Gralle
- Instituto de Bioquímica Médica, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | | |
Collapse
|
255
|
Kimura N, Imamura O, Ono F, Terao K. Aging attenuates dynactin–dynein interaction: Down-regulation of dynein causes accumulation of endogenous tau and amyloid precursor protein in human neuroblastoma cells. J Neurosci Res 2007; 85:2909-16. [PMID: 17628503 DOI: 10.1002/jnr.21408] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Impaired axonal transport may promote pathogenesis in neurodegenerative disorders, such as Alzheimer's disease (AD). We previously showed that tau, amyloid precursor protein (APP), and intracellular amyloid beta-protein (Abeta) accumulate in the nerve-ending fraction of aged monkey brains, perhaps because of impaired axonal transport. In the present study, we assessed age-related changes of axonal transport motor proteins in aged monkey brains. Western blotting showed that kinesin, dynein, and dynactin (DYN) localizations dramatically changed with aging, and dynein level in nerve-ending fractions increased significantly. Coimmunoprecipitation analyses showed that DYN-dynein intermediate chain (DIC) interactions decreased, suggesting that age-related attenuation of this interaction may cause the impairment of dynein function. Moreover, RNAi-induced down-regulation of DIC in human neuroblastoma cells caused endogenous tau and APP to accumulate, and their subcellular localizations were also affected. Our findings suggest that aging attenuates DYN-DIC interaction, representing one of the risk factors for age-related impaired dynein function and even for accumulation of disease proteins.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Ibaraki, Japan.
| | | | | | | |
Collapse
|
256
|
Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng H, Fridkin M, Youdim MBH. Therapeutic targets and potential of the novel brain- permeable multifunctional iron chelator?monoamine oxidase inhibitor drug, M-30, for the treatment of Alzheimer's disease. J Neurochem 2007; 100:490-502. [PMID: 17144902 DOI: 10.1111/j.1471-4159.2006.04258.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Novel therapeutic approaches for the treatment of neurodegenerative disorders comprise drug candidates designed specifically to act on multiple CNS targets. We have synthesized a multifunctional non-toxic, brain permeable iron chelator drug, M-30, possessing propargyl monoamine oxidase (MAO) inhibitory neuroprotective and iron-chelating moieties, from our prototype iron chelator VK-28. In the present study M-30 was shown to possess a wide range of pharmacological activities, including pro-survival neurorescue effects, induction of neuronal differentiation and regulation of amyloid precursor protein (APP) and beta-amyloid (Abeta) levels. M-30 was found to decrease apoptosis of SH-SY5Y neuroblastoma cells in a neurorescue, serum deprivation model, via reduction of the pro-apoptotic proteins Bad and Bax, and inhibition of the apoptosis-associated phosphorylated H2A.X protein (Ser 139) and caspase 3 activation. In addition, M-30 induced the outgrowth of neurites, triggered cell cycle arrest in G(0)/G(1) phase and enhanced the expression of growth associated protein-43. Furthermore, M-30 markedly reduced the levels of cellular APP and beta-C-terminal fragment (beta-CTF) and the levels of the amyloidogenic Abeta peptide in the medium of SH-SY5Y cells and Chinese hamster ovary cells stably transfected with the APP 'Swedish' mutation. Levels of the non-amyloidogenic soluble APPalpha and alpha-CTF in the medium and cell lysate respectively were coordinately increased. These properties, together with its brain selective MAO inhibitory and propargylamine- dependent neuroprotective effects, suggest that M-30 might serve as an ideal drug for neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases, in which oxidative stress and iron dysregulation have been implicated.
Collapse
Affiliation(s)
- Yael Avramovich-Tirosh
- Eve Topf Centers of Excellence, Technion-Rappaport Family Faculty of Medicine and Department of Pharmacology, Haifa, Israel
| | | | | | | | | | | |
Collapse
|
257
|
Tian X, Maftei M, Kohlmann M, Allinquant B, Przybylski M. Differential epitope identification of antibodies against intracellular domains of alzheimer's amyloid precursor protein using high resolution affinity-mass spectrometry. Subcell Biochem 2007; 43:339-354. [PMID: 17953402 DOI: 10.1007/978-1-4020-5943-8_16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Several polypeptides comprising the carboxy-terminal domain of the 1-amyloid precursor protein (cAPP) were prepared by solid phase peptide synthesis, and employed as antigens for the determination of the epitopes recognised by anti-cAPP antibodies. Selective proteolytic epitope-excision and -extraction on the immobilised immune complexes, in combination with high resolution Fourier transform ion cyclotron resonance mass spectrometry (FTICR-MS) were used as major methods for epitope identification. The epitope recognised by a polyclonal anti-cAPP antibody (36-BO) was identified as APP(727-737), a sequence close to the APP transmembrane region. In contrast, the epitope recognised by a monoclonal anti-cAPP antibody (Jonas-mAb) was identified at APP(740-747) to be located more remote from the transmembrane region. The two adjacent, yet distinct epitopes recognised by two different antibodies should provide efficient tools for (i), molecular diagnostic applications, and (ii), the study of intracellular processing pathways of APP relevant to Alzheimer's disease, utilising suitable mass spectrometric and molecular imaging approaches.
Collapse
Affiliation(s)
- Xiaodan Tian
- Department of Chemistry, Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, University of Konstanz, Germany
| | | | | | | | | |
Collapse
|
258
|
Mandel S, Weinreb O, Reznichenko L, Kalfon L, Amit T. Green tea catechins as brain-permeable, non toxic iron chelators to "iron out iron" from the brain. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2006:249-57. [PMID: 17447435 DOI: 10.1007/978-3-211-33328-0_26] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Evidence to link abnormal metal (iron, copper and zinc) metabolism and handling with Parkinson's and Alzheimer's diseases pathology has frequently been reported. The capacity of free iron to enhance and promote the generation of toxic reactive oxygen radicals has been discussed numerous times. Metal chelation has the potential to prevent iron-induced oxidative stress and aggregation of alpha-synuclein and beta-amyloid peptides. The efficacy of iron chelators depends on their ability to penetrate the subcellular compartments and cellular membranes where iron dependent free radicals are generated. Thus, natural, non-toxic, brain permeable neuroprotective drugs, are preferentially advocated for "ironing out iron" from those brain areas where it preferentially accumulates in neurodegenerative diseases. This review will discuss the most recent findings from in vivo and in vitro studies concerning the transitional metal (iron and copper) chelating property of green tea, and its major polyphenol, (-)-epigallocatechin-3-gallate with respect to their potential for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- S Mandel
- Eve Topf and US NPF Centers for Neurodegenerative Diseases and Department of Pharmacology, Faculty of Medicine, Technion, Haifa, Israel.
| | | | | | | | | |
Collapse
|
259
|
Hoareau C, Borrell V, Soriano E, Krebs MO, Prochiantz A, Allinquant B. Amyloid precursor protein cytoplasmic domain antagonizes reelin neurite outgrowth inhibition of hippocampal neurons. Neurobiol Aging 2006; 29:542-53. [PMID: 17169463 DOI: 10.1016/j.neurobiolaging.2006.11.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 10/09/2006] [Accepted: 11/13/2006] [Indexed: 01/12/2023]
Abstract
The function of the amyloid precursor protein (APP), a key molecule in Alzheimer's disease (AD) remains unknown. Among the proteins that interact with the APP cytoplasmic domain in vitro and in heterologous systems is Disabled-1, a signaling molecule of the reelin pathway. The physiological consequence of this interaction is unknown. Here we used an in vitro model of hippocampal neurons grown on a reelin substrate that inhibits neurite outgrowth. Our results show that an excess of APP cytoplasmic domain internalized by a cell permeable peptide, is able to antagonize the neurite outgrowth inhibition of reelin. The APP cytoplasmic domain binds Disabled-1 and retains it in the cytoplasm, preventing it from reaching the plasma membrane and sequesters tyrosine phosphorylated Disabled-1, both of which disrupt reelin signaling. In the context of AD, increased formation of APP cytoplasmic domain in the cytosol released after cleavage of the A beta peptide, could then inhibit reelin signaling pathway in the hippocampus and thus influence synaptic plasticity.
Collapse
Affiliation(s)
- C Hoareau
- INSERM U796, Centre Paul Broca, 2 ter rue d'Alésia, 75014 Paris, France
| | | | | | | | | | | |
Collapse
|
260
|
Meckler X, Bertandeau E, McNiven MA, Allinquant B, Hémar A. The cytosolic domain of APP induces the relocalization of dynamin 3 in hippocampal neurons. Eur J Neurosci 2006; 24:2439-43. [PMID: 17100832 DOI: 10.1111/j.1460-9568.2006.05141.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amyloid precursor protein (APP) has been the subject of intense research to uncover its implication in Alzheimer's disease. Its physiological function is, however, still poorly understood. Herein, we investigated its possible influence on the development of cultured hippocampal neurons. A peptide corresponding to the APP intracellular domain linked to a cell-penetrating peptide was used to alter the interactions of APP with its cytosolic partners. This treatment promoted the concentration of the cytosolic GTPase dynamin 3 (Dyn3) in neurite segments when most untreated cells displayed a homogenous punctate distribution of Dyn3. The Dyn3-labelled segments were excluded from those revealed by APP staining after aldehyde fixation. Interestingly, after aldehyde fixation MAP2 also labelled segments excluded from APP-stained segments. Thus APP is also a marker for the spacing pattern of neurites demonstrated by Taylor & Fallon (2006)J. Neurosci., 26, 1154-4463.
Collapse
Affiliation(s)
- X Meckler
- Physiologie Cellulaire de la Synapse, UMR5091 CNRS, Université Victor Segalen Bordeaux 2, 146 rue Léo Saignat, 33077 Bordeaux Cedex, France
| | | | | | | | | |
Collapse
|
261
|
Senechal Y, Larmet Y, Dev KK. Unraveling in vivo functions of amyloid precursor protein: insights from knockout and knockdown studies. NEURODEGENER DIS 2006; 3:134-47. [PMID: 16954700 DOI: 10.1159/000094772] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The amyloid precursor protein (APP) is a widely expressed transmembrane protein that is cleaved to generate Abeta peptides in the central nervous system and is a key player in the pathogenesis of Alzheimer's disease. The precise biological functions of APP still remain unclear although various roles have been proposed. While a commonly accepted model argues that Abeta peptides are the cause of onset and early pathogenesis of Alzheimer's disease, recent discussions challenge this 'Abeta hypothesis' and suggest a direct role for APP in this neurodegenerative disease. Loss-of-function studies are an efficient way to elucidate the role of proteins and concurrently a variety of in vitro and in vivo studies has been performed for APP where protein levels have been downregulated and functional consequences monitored. Complete disruption of APP gene expression has been achieved by the generation of APP knockout animal models. Further knockdown studies using antisense and RNA interference have allowed scientists to reduce APP expression levels and have opened new avenues to explore the physiological roles of APP. In the present review, we focus on knockout and knockdown approaches that have provided insights into the physiological functions of APP and discuss their advantages and drawbacks.
Collapse
Affiliation(s)
- Yann Senechal
- Neuroscience Research, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | |
Collapse
|
262
|
Satpute-Krishnan P, DeGiorgis JA, Conley MP, Jang M, Bearer EL. A peptide zipcode sufficient for anterograde transport within amyloid precursor protein. Proc Natl Acad Sci U S A 2006; 103:16532-7. [PMID: 17062754 PMCID: PMC1621108 DOI: 10.1073/pnas.0607527103] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fast anterograde transport of membrane-bound organelles delivers molecules synthesized in the neuronal cell body outward to distant synapses. Identification of the molecular "zipcodes" on organelles that mediate attachment and activation of microtubule-based motors for this directed transport is a major area of inquiry. Here we identify a short peptide sequence (15 aa) from the cytoplasmic C terminus of amyloid precursor protein (APP-C) sufficient to mediate the anterograde transport of peptide-conjugated beads in the squid giant axon. APP-C beads travel at fast axonal transport rates (0.53 mum/s average velocity, 0.9 mum/s maximal velocity) whereas beads coupled to other peptides coinjected into the same axon remain stationary at the injection site. This transport appears physiologic, because it mimics behavior of endogenous squid organelles and of beads conjugated to C99, a polypeptide containing the full-length cytoplasmic domain of amyloid precursor protein (APP). Beads conjugated to APP lacking the APP-C domain are not transported. Coinjection of APP-C peptide reduces C99 bead motility by 75% and abolishes APP-C bead motility, suggesting that the soluble peptide competes with protein-conjugated beads for axoplasmic motor(s). The APP-C domain is conserved (13/15 aa) from squid to human, and peptides from either squid or human APP behave similarly. Thus, we have identified a conserved peptide zipcode sufficient to direct anterograde transport of exogenous cargo and suggest that one of APP's roles may be to recruit and activate axonal machinery for endogenous cargo transport.
Collapse
Affiliation(s)
- Prasanna Satpute-Krishnan
- *Department of Pathology and Laboratory Medicine, Brown University Medical School, Providence, RI 02912
- Marine Biological Laboratory, Woods Hole, MA 02543; and
| | - Joseph A. DeGiorgis
- Marine Biological Laboratory, Woods Hole, MA 02543; and
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Michael P. Conley
- *Department of Pathology and Laboratory Medicine, Brown University Medical School, Providence, RI 02912
- Marine Biological Laboratory, Woods Hole, MA 02543; and
| | - Marcus Jang
- *Department of Pathology and Laboratory Medicine, Brown University Medical School, Providence, RI 02912
- Marine Biological Laboratory, Woods Hole, MA 02543; and
| | - Elaine L. Bearer
- *Department of Pathology and Laboratory Medicine, Brown University Medical School, Providence, RI 02912
- Marine Biological Laboratory, Woods Hole, MA 02543; and
- To whom correspondence should be addressed at:
Brown University Medical School, 70 Ship Street, G-E527, Providence, RI 02912. E-mail:
| |
Collapse
|
263
|
Kins S, Lauther N, Szodorai A, Beyreuther K. Subcellular Trafficking of the Amyloid Precursor Protein Gene Family and Its Pathogenic Role in Alzheimer’s Disease. NEURODEGENER DIS 2006; 3:218-26. [PMID: 17047360 DOI: 10.1159/000095259] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Changes in the intracellular transport of amyloid precursor protein (APP) affect the extent to which APP is exposed to alpha- or beta-secretase in a common subcellular compartment and therefore directly influence the degree to which APP undergoes the amyloidogenic pathway leading to generation of beta-amyloid. As the presynaptic regions of neurons are thought to be the main source of beta-amyloid in the brain, attention has been focused on axonal APP trafficking. APP is transported along axons by a fast, kinesin-dependent anterograde transport mechanism. Despite the wealth of in vivo and in vitro data that have accumulated regarding the connection of APP to kinesin transport, it is not yet clear if APP is coupled to its specific motor protein via an intracellular interaction partner, such as the c-Jun N-terminal kinase-interacting protein, or by yet another unknown molecular mechanism. The cargo proteins that form a functional complex with APP are also unknown. Due to the long lifespan, and vast extent, of neurons, in particular axons, neurons are highly sensitive to changes in subcellular transport. Recent in vitro and in vivo studies have shown that variations in APP or tau affect mitochondrial and synaptic vesicle transport. Further, it was shown that this axonal dysfunction might lead to impaired synaptic plasticity, which is crucial for neuronal viability and function. Thus, changes in APP and tau expression may cause perturbed axonal transport and changes in APP processing, contributing to cognitive decline and neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Stefan Kins
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany.
| | | | | | | |
Collapse
|
264
|
Grimm MOW, Tschäpe JA, Grimm HS, Zinser EG, Hartmann T. Altered membrane fluidity and lipid raft composition in presenilin-deficient cells. Acta Neurol Scand 2006; 185:27-32. [PMID: 16866908 DOI: 10.1111/j.1600-0404.2006.00682.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pathology of Alzheimer's disease is closely connected with lipid metabolism. Processing of amyloid precursor protein (APP) is sensitive to membrane alterations in levels of cholesterol and gangliosides. As cholesterol and gangliosides are major components of rafts and BACE I and gamma-secretase are supposed to be localized to rafts there might be a yet unknown biological function underlying this connection. Increasing evidence shows a close connection between cholesterol homeostasis and APP processing and Abeta production respectively. We measured membrane fluidity by anisotropy determination, isolated detergent resistant membrane (DRM) fractions from membrane preparations and determined cholesterol content of these fractions by a coupled enzymatic assay. We found membrane fluidity to be changed in mouse embryonic fibroblasts (MEF) PS1/2 -/- along with altered cholesterol content in DRM fraction of these cells. In addition, total ganglioside levels were enhanced in absence of presenilin (PS).
Collapse
Affiliation(s)
- M O W Grimm
- Centre for Molecular Biology Heidelberg, Germany
| | | | | | | | | |
Collapse
|
265
|
Xue S, Jia L, Jia J. Hypoxia and reoxygenation increased BACE1 mRNA and protein levels in human neuroblastoma SH-SY5Y cells. Neurosci Lett 2006; 405:231-5. [PMID: 16901640 DOI: 10.1016/j.neulet.2006.07.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Revised: 06/30/2006] [Accepted: 07/06/2006] [Indexed: 10/24/2022]
Abstract
Ischemic cerebrovascular diseases, usually involved in hypoxia and reoxygenation, have been reported to increase the risk of dementia such as Alzheimer's disease (AD). beta-site amyloid protein precursor (APP)-cleaving enzymes (BACE1) have been identified to participate in the secretion of beta-amyloid peptides (Abeta), and its expressive alteration would contribute to the AD neuropathology. We have investigated the effect of hypoxia (0% O(2), 24h) and reoxygenation (0h, 12h and 24h after 24h hypoxia) on BACE1 mRNA and protein levels in human neuroblastoma SH-SY5Y cells. At the same time, we also examined the effect of hypoxia and reoxygenation on APP mRNA and protein levels. We demonstrated that hypoxia and reoxygenation did not alter APP mRNA and protein level, However compared to those of controls, BACE1 mRNA levels were up-regulated by 31.5% (P=0.028) and 35.1% (P=0.005) at 12h and 24h and the protein levels increased to 22%(P=0.021), 42% (P=0.000) and 51.5% (P=0.000) at 0h, 12h and 24h after reoxygenation, respectively. Thus by up-regulating of BACE1 mRNA and protein level in the neuronal cell, hypoxia may be a linkage in the pathophysiology between cerebravascular diseases and AD.
Collapse
Affiliation(s)
- Sufang Xue
- Department of Neurology, Xuanwu Hospital of the Capital University of Medical Sciences, Neurodegenerative Lab of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | | | | |
Collapse
|
266
|
Pastorino L, Lu KP. Pathogenic mechanisms in Alzheimer's disease. Eur J Pharmacol 2006; 545:29-38. [PMID: 16904664 DOI: 10.1016/j.ejphar.2006.06.078] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 06/23/2006] [Accepted: 06/27/2006] [Indexed: 01/22/2023]
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.
Collapse
Affiliation(s)
- Lucia Pastorino
- Cancer Biology Program, Beth Israel Deaconess Medical Center, Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA 02115, USA.
| | | |
Collapse
|
267
|
Transcriptional and translational regulation of BACE1 expression--implications for Alzheimer's disease. Prog Neurobiol 2006; 79:95-111. [PMID: 16904810 DOI: 10.1016/j.pneurobio.2006.06.001] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Revised: 06/08/2006] [Accepted: 06/09/2006] [Indexed: 01/07/2023]
Abstract
The proteolytical processing of the amyloid precursor protein (APP) gives rise to beta-amyloid peptides, which accumulate in brains of Alzheimer's disease (AD) patients. Different soluble or insoluble higher molecular weight forms of beta-amyloid peptides have been postulated to trigger a complex pathological cascade that may cause synaptic dysfunction, inflammatory processes, neuronal loss, cognitive impairment, and finally the onset of the disease. The generation of beta-amyloid peptides requires the proteolytical cleavage of APP by an aspartyl protease named beta-site APP-cleaving enzyme 1 (BACE1). The expression and enzymatic activity of BACE1 are increased in brains of AD patients. Here we discuss the importance of a number of recently identified transcription factors as well as post-transcriptional modifications and activation of intracellular signaling molecules for the regulation of BACE1 expression in brain. Importantly, some of these factors are known to be involved in the inflammatory and chronic stress responses of the brain, which are compromised during aging. Moreover, recent evidence indicates that beneficial effects of non-steriodal anti-inflammatory drugs on the progression of AD are mediated--at least in part--by effects on the peroxisome proliferator-activated receptor-gamma response element present in the BACE1 promoter. The identification of the cell type-specific expression and activation of NF-kappaB, Sp1 and YY1 transcription factors may provide a basis to specifically interfere with BACE1 expression and, thereby, to lower the concentrations of beta-amyloid peptides, which may prevent neuronal cell loss and cognitive decline in AD patients.
Collapse
|
268
|
Nistor M, Don M, Parekh M, Sarsoza F, Goodus M, Lopez G, Kawas C, Leverenz J, Doran E, Lott I, Hill M, Head E. Alpha- and beta-secretase activity as a function of age and beta-amyloid in Down syndrome and normal brain. Neurobiol Aging 2006; 28:1493-506. [PMID: 16904243 PMCID: PMC3375834 DOI: 10.1016/j.neurobiolaging.2006.06.023] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 05/09/2006] [Accepted: 06/21/2006] [Indexed: 11/22/2022]
Abstract
Aged individuals with Down syndrome (DS) develop Alzheimer's disease (AD) neuropathology by the age of 40 years. The purpose of the current study was to measure age-associated changes in APP processing in 36 individuals with DS (5 months-69 years) and in 26 controls (5 months-100 years). Alpha-secretase significantly decreased with age in DS, particularly in cases over the age of 40 years and was stable in controls. The levels of C-terminal fragments of APP reflecting alpha-secretase processing (CTF-alpha) decreased with age in both groups. In both groups, there was significant increase in beta-secretase activity with age. CTF-beta remained constant with age in controls suggesting compensatory increases in turnover/clearance mechanisms. In DS, young individuals had the lowest CTF-beta levels that may reflect rapid conversion of beta-amyloid (Abeta) to soluble pools or efficient CTF-beta clearance mechanisms. Treatments to slow or prevent AD in the general population targeting secretase activity may be more efficacious in adults with DS if combined with approaches that enhance Abeta degradation and clearance.
Collapse
Affiliation(s)
- M. Nistor
- Institute for Brain Aging & Dementia, CA, United States
| | - M. Don
- Institute for Brain Aging & Dementia, CA, United States
| | - M. Parekh
- Institute for Brain Aging & Dementia, CA, United States
| | - F. Sarsoza
- Institute for Brain Aging & Dementia, CA, United States
| | - M. Goodus
- Institute for Brain Aging & Dementia, CA, United States
- Department of Neurology, University of California, 1259 Gillespie NRF, Irvine, CA 92697-4540, United States
| | - G.E. Lopez
- Institute for Brain Aging & Dementia, CA, United States
- Department of Neurology, University of California, 1259 Gillespie NRF, Irvine, CA 92697-4540, United States
| | - C. Kawas
- Institute for Brain Aging & Dementia, CA, United States
- Department of Neurology, University of California, 1259 Gillespie NRF, Irvine, CA 92697-4540, United States
| | - J. Leverenz
- Neurology and Psychiatry and Behavioral Sciences, University of Washington Seattle, WA 98108, United States
| | - E. Doran
- Institute for Brain Aging & Dementia, CA, United States
| | - I.T. Lott
- Institute for Brain Aging & Dementia, CA, United States
- Department of Neurology, University of California, 1259 Gillespie NRF, Irvine, CA 92697-4540, United States
| | - M. Hill
- Institute for Brain Aging & Dementia, CA, United States
- Department of Neurology, University of California, 1259 Gillespie NRF, Irvine, CA 92697-4540, United States
| | - E. Head
- Institute for Brain Aging & Dementia, CA, United States
- Department of Neurology, University of California, 1259 Gillespie NRF, Irvine, CA 92697-4540, United States
- Corresponding author. Tel.: +1 949 824 8700; fax: +1 949 824 2071. (E. Head)
| |
Collapse
|
269
|
Muresan Z, Muresan V. Neuritic deposits of amyloid-beta peptide in a subpopulation of central nervous system-derived neuronal cells. Mol Cell Biol 2006; 26:4982-97. [PMID: 16782885 PMCID: PMC1489158 DOI: 10.1128/mcb.00371-06] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 04/04/2006] [Accepted: 04/18/2006] [Indexed: 11/20/2022] Open
Abstract
Our goal is to understand the pathogenesis of amyloid-beta (Abeta) deposition in the Alzheimer's disease (AD) brain. We established a cell culture system where central nervous system-derived neuronal cells (CAD cells) produce and accumulate within their processes large amounts of Abeta peptide, similar to what is believed to occur in brain neurons, in the initial phases of AD. Using this system, we show that accumulation of Abeta begins within neurites, prior to any detectable signs of neurodegeneration or abnormal vesicular transport. Neuritic accumulation of Abeta is restricted to a small population of neighboring cells that express normal levels of amyloid-beta precursor protein (APP) but show redistribution of BACE1 to the processes, where it colocalizes with Abeta and markers of late endosomes. Consistently, cells that accumulate Abeta appear in isolated islets, suggesting their clonal origin from a few cells that show a propensity to accumulate Abeta. These results suggest that Abeta accumulation is initiated in a small number of neurons by intracellular determinants that alter APP metabolism and lead to Abeta deposition and neurodegeneration. CAD cells appear to recapitulate the biochemical processes leading to Abeta deposition, thus providing an experimental in vitro system for studying the molecular pathobiology of AD.
Collapse
Affiliation(s)
- Zoia Muresan
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106-4970, USA
| | | |
Collapse
|
270
|
Tate BA, Mathews PM. Targeting the role of the endosome in the pathophysiology of Alzheimer's disease: a strategy for treatment. ACTA ACUST UNITED AC 2006; 2006:re2. [PMID: 16807486 DOI: 10.1126/sageke.2006.10.re2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Membrane-bound endosomal vesicles play an integral role in multiple cellular events, including protein processing and turnover, and often critically regulate the cell-surface availability of receptors and other plasma membrane proteins in many different cell types. Neurons are no exception, being dependent on endosomal function for housekeeping and synaptic events. Growing evidence suggests a link between neuronal endosomal function and Alzheimer's disease (AD) pathophysiology. Endosomal abnormalities invariably occur within neurons in AD brains, and endocytic compartments are one likely site for the production of the pathogenic beta-amyloid peptide (Abeta), which accumulates within the brain during the disease and is generated by proteolytic processing of the amyloid precursor protein (APP). The enzymes and events involved in APP processing are appealing targets for therapeutic agents aimed at slowing or reversing the pathogenesis of AD. The neuronal endosome may well prove to be the intracellular site of action for inhibitors of beta-amyloidogenic APP processing. We present here the view that knowledge of the endosomal system in the disease can guide drug discovery of AD therapeutic agents.
Collapse
Affiliation(s)
- Barbara A Tate
- CNS Discovery, Global Research & Development, Pfizer Inc., Groton, CT 06234, USA.
| | | |
Collapse
|
271
|
Ledesma MD, Dotti CG. Amyloid excess in Alzheimer's disease: What is cholesterol to be blamed for? FEBS Lett 2006; 580:5525-32. [PMID: 16814780 DOI: 10.1016/j.febslet.2006.06.038] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Revised: 06/13/2006] [Accepted: 06/14/2006] [Indexed: 10/24/2022]
Abstract
A link between alterations in cholesterol homeostasis and Alzheimer's disease (AD) is nowadays widely accepted. However, the molecular mechanism/s underlying such link remain unclear. Numerous experimental evidences support the view that changes in neuronal membrane cholesterol levels and/or subcellular distribution determine the aberrant accumulation of the amyloid peptide in the disease. Still, this view comes from rather contradictory data supporting the existence of either high or low brain cholesterol content. This is of particular concern considering that therapeutical strategies aimed to reduce cholesterol levels are already being tested in humans. Here, we review the molecular mechanisms proposed and discuss the perspectives they open.
Collapse
Affiliation(s)
- Maria Dolores Ledesma
- Department of Molecular and Developmental Genetics, Flanders Interuniversity Institute of Biotechnology, VIB11, and Catholic University of Leuven, Campus Gasthuisberg, Heerstraat 49, 3000 Leuven, Belgium.
| | | |
Collapse
|
272
|
Gong Y, Meyer EM, Meyers CA, Klein RL, King MA, Hughes JA. Memory-related deficits following selective hippocampal expression of Swedish mutation amyloid precursor protein in the rat. Exp Neurol 2006; 200:371-7. [PMID: 16780838 DOI: 10.1016/j.expneurol.2006.02.136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 02/03/2006] [Accepted: 02/22/2006] [Indexed: 10/24/2022]
Abstract
The gene encoding for the Swedish double mutation (K595N/M596L) of amyloid precursor protein (APP695Swe) was expressed bilaterally in adult rat hippocampus to determine its long-term effects on memory-related behavior as well as amyloid deposition. Recombinant adeno-associated viral serotype 2 (rAAV2) vectors were injected that contained either non-expressing DNA or cDNA encoding for APP695Swe under control of a chicken beta actin/cytomegalovirus promoter/enhancer. Immunolabeling human APP with the antibody 6E10 was observed throughout the cytoplasm of aspiny and, to a lesser extent, spine-bearing hippocampal neurons 6 and 12 months post-injection of the APP695Swe but not control vector. Abeta1-42 immunolabeling was identified in unusual immunoreactive objects within the hilus of the dentate gyrus and in the granule cell layer, proximal to the injection site. At 12 months post-transduction, rats that received the APP695Swe gene also demonstrated significant deficits in the acquisition and probe components of the spatial-memory-related Morris water task compared to control animals. These behavioral deficits occurred in the absence of any amyloid plaques, gliosis, or FluoroJade labeling of dying neurons. In conclusion, prolonged and localized APP695Swe expression in hippocampal neurons is sufficient to produce memory deficits without plaque formation or neuronal loss.
Collapse
Affiliation(s)
- Yan Gong
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, 32610, USA
| | | | | | | | | | | |
Collapse
|
273
|
Marshall AJ, Rattray M, Vaughan PFT. Chronic hypoxia in the human neuroblastoma SH-SY5Y causes reduced expression of the putative alpha-secretases, ADAM10 and TACE, without altering their mRNA levels. Brain Res 2006; 1099:18-24. [PMID: 16762326 DOI: 10.1016/j.brainres.2006.05.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2005] [Revised: 04/03/2006] [Accepted: 05/03/2006] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease is more frequent following an ischemic or hypoxic episode, with levels of beta-amyloid peptides elevated in brains from patients. Similar increases are found after experimental ischemia in animals. It is possible that increased beta-amyloid deposition arises from alterations in amyloid precursor protein (APP) metabolism, indeed, we have shown that exposing cells of neuronal origin to chronic hypoxia decreased the secretion of soluble APP (sAPPalpha) derived by action of alpha-secretase on APP, coinciding with a decrease in protein levels of ADAM10, a disintegrin metalloprotease which is thought to be the major alpha-secretase. In the current study, we extended those observations to determine whether the expression of ADAM10 and another putative alpha-secretase, TACE, as well as the beta-secretase, BACE1 were regulated by chronic hypoxia at the level of protein and mRNA. Using Western blotting and RT-PCR, we demonstrate that after 48 h chronic hypoxia, such that sAPPalpha secretion is decreased by over 50%, protein levels of ADAM10 and TACE and by approximately 60% and 40% respectively with no significant decrease in BACE1 levels. In contrast, no change in the expression of the mRNA for these proteins could be detected. Thus, we conclude that under CH the level of the putative alpha-secretases, ADAM10 and TACE are regulated by post-translational mechanisms, most probably proteolysis, rather than at the level of transcription.
Collapse
Affiliation(s)
- Amy J Marshall
- King's College London, Wolfson Centre for Age-related Diseases, School of Biomedical and Health Sciences, Wolfson Wing, Guy's Campus, UK
| | | | | |
Collapse
|
274
|
Kelicen P, Nordberg A. Feedback regulation of SREBP and aromatase in A beta(25-35)-supplemented human neuroblastoma cells. Cell Mol Neurobiol 2006; 26:225-35. [PMID: 16767510 PMCID: PMC11520627 DOI: 10.1007/s10571-006-9060-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Accepted: 03/14/2006] [Indexed: 11/27/2022]
Abstract
1. The analogies between the processing of amyloid precursor protein (APP) and other transmembrane sterol regulatory element binding proteins (SREBPs) inspired us to conduct further studies on whether beta-amyloid (Abeta) affects aromatase by interacting with APP and SREBP. 2. In this study, cultured human neuroblastoma cells (SHSY-5Y) were incubated in experimental media (media without FBS, the main cholesterol source) in the presence or absence of Abeta (1 microM) for 24 h. 3. Cellular extracts were subjected to immunoblot analysis using anti-APP, anti-aromatase and anti-SREBP-1. In these cell lines, we detected aromatase (55 kDa), SREBP cleavage product (68 kDa) and APP precursor (100-95 kDa) and cleavage product (60 kDa) by immunoblotting. Aromatase and SREBP levels were elevated in the cells incubated 24 h in experimental media and were attenuated in Abeta-supplemented experimental media. 4. The disturbance of cholesterol homeostasis appears to be an important factor in the pathogenesis of Alzheimer's disease. These findings may have important implications for understanding the mechanisms of the aromatase enzyme gene in disease states such as Alzheimer's.
Collapse
Affiliation(s)
- Pelin Kelicen
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, 06100 Sihhiye, Ankara, Turkey.
| | | |
Collapse
|
275
|
Wirths O, Multhaup G, Czech C, Feldmann N, Blanchard V, Tremp G, Beyreuther K, Pradier L, Bayer TA. Intraneuronal APP/A beta trafficking and plaque formation in beta-amyloid precursor protein and presenilin-1 transgenic mice. Brain Pathol 2006; 12:275-86. [PMID: 12146796 PMCID: PMC8095864 DOI: 10.1111/j.1750-3639.2002.tb00442.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neuropil deposition of beta-amyloid peptides A beta40 and A beta42 is believed to be the key event in the neurodegenerative processes of Alzheimer's disease (AD). Since A beta seems to carry a transport signal that is required for axonal sorting of its precursor beta-amyloid precursor protein (APP), we studied the intraneuronal staining profile of A beta peptides in a transgenic mouse model expressing human mutant APP751 (KM670/671NL and V7171) and human mutant presenilin-1 (PS-1 M146L) in neurons. Using surface plasmon resonance we analyzed the A beta antibodies and defined their binding profile to APP, A beta40 and A beta42. Immunohistochemical staining revealed that intraneuronal A beta40 and A beta42 staining preceded plaque deposition, which started at 3 months of age. A beta was observed in the somatodendritic and axonal compartments of many neurons. Interestingly, the striatum, which lacks transgenic APP expression harbored many plaques at 10 months of age. This is most likely due to an APP/A beta transport problem and may be a model region to study APP/A beta trafficking as an early pathological event.
Collapse
Affiliation(s)
- Oliver Wirths
- Department of Psychiatry, University of Bonn Medical Center, Germany
| | - Gerd Multhaup
- Center for Molecular Biology University of Heidelberg, Germany
| | | | - Nicole Feldmann
- Department of Psychiatry, University of Bonn Medical Center, Germany
| | | | | | | | | | - Thomas A. Bayer
- Department of Psychiatry, University of Bonn Medical Center, Germany
| |
Collapse
|
276
|
Chen CD, Oh SY, Hinman JD, Abraham CR. Visualization of APP dimerization and APP-Notch2 heterodimerization in living cells using bimolecular fluorescence complementation. J Neurochem 2006; 97:30-43. [PMID: 16515557 DOI: 10.1111/j.1471-4159.2006.03705.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We previously demonstrated that the amyloid precursor protein (APP) interacts with Notch receptors. Here, we confirmed the APP/Notch1 endogenous interaction in embryonic day 17 rat brain tissue, suggesting the interaction was not as a result of over-expression artifacts. To investigate potential homodimeric and heterodimeric interactions of APP and Notch2 (N2), we have visualized the subcellular localization of the APP/N2 complexes formed in living cells using bimolecular fluorescence complementation (BiFC) analysis. BiFC was accomplished by fusing the N-terminal fragment or the C-terminal fragment of yellow fluorescent protein (YFP) to APP, N2, and a C-terminally truncated form of N2. When expressed in COS-7 cells, these tagged proteins alone did not produce a fluorescent signal. The tagged APP homodimer produced a weak fluorescent signal, while neither full-length N2, nor a truncated N2 alone, produced a visible signal, suggesting that N2 receptors do not form homodimers. The strongest fluorescent signal was obtained with co-expression of the C-terminal fragment of YFP fused to APP and the N-terminal fragment of YFP fused to the truncated form of N2. This heterodimer localized to plasma membrane, endoplasmic reticulum (ER), Golgi and other compartments. The results were confirmed and quantified by flow cytometry. The BiFC method of specifically visualizing APP/Notch interactions can be applied to study APP and Notch signaling during development, aging and neurodegeneration.
Collapse
Affiliation(s)
- Ci-Di Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
277
|
Reznichenko L, Amit T, Zheng H, Avramovich-Tirosh Y, Youdim MBH, Weinreb O, Mandel S. Reduction of iron-regulated amyloid precursor protein and beta-amyloid peptide by (-)-epigallocatechin-3-gallate in cell cultures: implications for iron chelation in Alzheimer's disease. J Neurochem 2006; 97:527-36. [PMID: 16539659 DOI: 10.1111/j.1471-4159.2006.03770.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Brain iron dysregulation and its association with amyloid precursor protein (APP) plaque formation are implicated in Alzheimer's disease (AD) pathology and so iron chelation could be considered a rational therapeutic strategy for AD. Here we analyzed the effect of the main polyphenol constituent of green tea, (-)-epigallocatechin-3-gallate (EGCG), which possesses metal-chelating and radical-scavenging properties, on the regulation of the iron metabolism-related proteins APP and transferrin receptor (TfR). EGCG exhibited potent iron-chelating activity comparable to that of the prototype iron chelator desferrioxamine, and dose dependently (1-10 microm) increased TfR protein and mRNA levels in human SH-SY5Y neuroblastoma cells. Both the immature and full-length cellular holo-APP were significantly reduced by EGCG, as shown by two-dimensional gel electrophoresis, without altering APP mRNA levels, suggesting a post-transcriptional action. Indeed, EGCG suppressed the translation of a luciferase reporter gene fused to the APP mRNA 5'-untranslated region, encompassing the APP iron-responsive element. The finding that Fe(2)SO(4) reversed the action of EGCG on APP and TfR proteins reinforces the likelihood that these effects are mediated through modulation of the intracellular iron pool. Furthermore, EGCG reduced toxic beta-amyloid peptide generation in Chinese hamster ovary cells overexpressing the APP 'Swedish' mutation. Thus, the natural non-toxic brain-permeable EGCG may provide a potential therapeutic approach for AD and other iron-associated disorders.
Collapse
Affiliation(s)
- L Reznichenko
- Eve Topf and US National Parkinson Foundation Centers for Neurodegenerative diseases and Department of Pharmacology, Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
278
|
Ahmad M, Takino T, Miyamori H, Yoshizaki T, Furukawa M, Sato H. Cleavage of Amyloid-β Precursor Protein (APP) by Membrane-Type Matrix Metalloproteinases. ACTA ACUST UNITED AC 2006; 139:517-26. [PMID: 16567416 DOI: 10.1093/jb/mvj054] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Amyloid-beta precursor protein (APP) was identified on expression cloning from a human placenta cDNA library as a gene product that modulates the activity of membrane-type matrix metalloproteinase-1 (MT1-MMP). Co-expression of MT1-MMP with APP in HEK293T cells induced cleavage and shedding of the APP ectodomain when co-expressed with APP adaptor protein Fe65. Among the MT-MMPs tested, MT3-MMP and MT5-MMP also caused efficient APP shedding. The recombinant APP protein was cleaved by MT3-MMP in vitro at the A463-M464, N579-M580, H622-S623, and H685-Q686 peptide bonds, which included a cleavage site within the amyloid beta peptide region known to produce a C-terminal fragment. The Swedish-type mutant of APP, which produces a high level of amyloid beta peptide, was more effectively cleaved by MT3-MMP than wild-type APP in both the presence and absence of Fe65; however, amyloid beta peptide production was not affected by MT3-MMP expression. Expression of MT3-MMP enhanced Fe65-dependent transactivation by APP fused to the Gal4 DNA-binding and transactivation domains. These results suggest that MT1-MMP, MT3-MMP and MT5-MMP should play an important role in the regulation of APP functions in tissues including the central nervous system.
Collapse
Affiliation(s)
- Munirah Ahmad
- Department of Molecular Virology and Oncology, Cancer Research Institute, Kanazawa University, Takara-machi, Ishikawa
| | | | | | | | | | | |
Collapse
|
279
|
Oh SY, Ellenstein A, Chen CD, Hinman JD, Berg EA, Costello CE, Yamin R, Neve RL, Abraham CR. Amyloid precursor protein interacts with notch receptors. J Neurosci Res 2006; 82:32-42. [PMID: 16118793 DOI: 10.1002/jnr.20625] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The amyloid precursor protein (APP) must fulfill important roles based on its sequence conservation from fly to human. Although multiple functions for APP have been proposed, the best-known role for this protein is as the precursor of Abeta peptide, a neurotoxic 39-43-amino acid peptide crucial to the pathogenesis of Alzheimer's disease. To investigate additional roles for APP with an eye toward understanding the molecular basis of the pleiotropic effects ascribed to APP, we isolated proteins that interacted with the plasma membrane isoform of APP. We employed a membrane-impermeable crosslinker to immobilize proteins binding to transmembrane APP in human embryonic kidney (HEK)293 cells expressing APP751 (HEK275) or rat embryonic day 18 primary neurons infected with a virus expressing APP. Notch2 was identified as a potential APP binding partner based on mass spectrometry analysis of APP complexes immunopurified from neurons. To confirm the interaction between Notch2 and APP, we carried out immunoprecipitation studies in HEK275 cells transiently expressing full-length Notch2 using Notch2 antibodies. The results indicated that APP and Notch2 interact in mammalian cells, and confirmed our initial findings. Interestingly, Notch1 also coimmunoprecipitated with APP, suggesting that APP and Notch family members may engage in intermolecular cross talk to modulate cell function. Finally, cotransfection of APP/CFP and Notch2/YFP into COS cells revealed that these two proteins colocalize on the plasma membrane. Intracellularly, however, although some APP and Notch molecules colocalize, others reside in distinct locations. The discovery of proteins that interact with APP may aid in the identification of new functions for APP.
Collapse
Affiliation(s)
- Sun Young Oh
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
280
|
Andersen OM, Schmidt V, Spoelgen R, Gliemann J, Behlke J, Galatis D, McKinstry WJ, Parker MW, Masters CL, Hyman BT, Cappai R, Willnow TE. Molecular Dissection of the Interaction between Amyloid Precursor Protein and Its Neuronal Trafficking Receptor SorLA/LR11. Biochemistry 2006; 45:2618-28. [PMID: 16489755 DOI: 10.1021/bi052120v] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
SorLA/LR11 is a sorting receptor that regulates the intracellular transport and processing of the amyloid precursor protein (APP) in neurons. SorLA/LR11-mediated binding results in sequestration of APP in the Golgi and in protection from processing into the amyloid-beta peptide (Abeta), the principal component of senile plaques in Alzheimer's disease (AD). To gain insight into the molecular mechanisms governing sorLA and APP interaction, we have dissected the respective protein interacting domains. Using a fluorescence resonance energy transfer (FRET) based assay of protein proximity, we identified binding sites in the extracellular regions of both proteins. Fine mapping by surface plasmon resonance analysis and analytical ultracentrifugation of recombinant APP and sorLA fragments further narrowed down the binding domains to the cluster of complement-type repeats in sorLA that forms a 1:1 stoichiometric complex with the carbohydrate-linked domain of APP. These data shed new light on the molecular determinants of neuronal APP trafficking and processing and on possible targets for intervention with senile plaque formation in patients with AD.
Collapse
Affiliation(s)
- Olav M Andersen
- Max-Delbrueck-Center for Molecular Medicine, Berlin, 13125 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
281
|
Kim JH, Kim HJ. Direct involvement of G protein alpha(q/11) subunit in regulation of muscarinic receptor-mediated sAPPalpha release. Arch Pharm Res 2006; 28:1275-81. [PMID: 16350855 DOI: 10.1007/bf02978212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The G(q/11) protein-coupled receptors, such as muscarinic (M1 & M3) receptors, have been shown to regulate the release of a soluble amyloid precursor protein (sAPPalpha) produced from alpha-secretase processing. However, there is no direct evidence for the precise characteristics of G proteins, and the signaling mechanism for the regulation of G(q/11) protein-coupled receptor-mediated sAPPalpha release is not clearly understood. This study examined whether the muscarinic receptor-mediated release of sAPPalpha is directly regulated by Galpha(q/11) proteins. The HEK293 cells were transiently cotransfected with muscarinic M3 receptors and a dominant-negative minigene construct of the G protein alpha subunit. The sAPPalpha release in the media was measured using an antibody specific for sAPP. The sAPPalpha release enhancement induced by muscarinic receptor stimulation was decreased by a G(q/11) minigene construct, whereas it was not blocked by a control minigene construct (the Galpha carboxy peptide in random order, Galpha(q)R) or Galpha(i) constructs. This indicated a direct role of the Galpha(q/11) protein in the regulation of muscarinic M3 receptor-mediated sAPPalpha release. We also investigated whether the transactivation of the epidermal growth factor receptor (EGFR) by a muscarinic agonist could regulate the sAPPalpha release in SH-SY5Y cells. Pretreatment of a specific EGFR kinase inhibitor, tyrophostin AG1478 (250 nM), blocked the EGF-stimulated sAPPalpha release, but did not block the oxoM-stimulated sAPPalpha release. This demonstrated that the transactivation of the EGFR by muscarinic receptor activation was not involved in the muscarinic receptor-mediated sAPPalpha release.
Collapse
Affiliation(s)
- Jin Hyoung Kim
- Division of Pharmaceutical Bioscience, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | | |
Collapse
|
282
|
Mandel S, Amit T, Reznichenko L, Weinreb O, Youdim MBH. Green tea catechins as brain-permeable, natural iron chelators-antioxidants for the treatment of neurodegenerative disorders. Mol Nutr Food Res 2006; 50:229-34. [PMID: 16470637 DOI: 10.1002/mnfr.200500156] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurodegeneration in Parkinson's, Alzheimer's, or other neurodegenerative diseases appears to be multifactorial, where a complex set of toxic reactions, including oxidative stress (OS), inflammation, reduced expression of trophic factors, and accumulation of protein aggregates, lead to the demise of neurons. One of the prominent pathological features is the abnormal accumulation of iron on top of the dying neurons and in the surrounding microglia. The capacity of free iron to enhance and promote the generation of toxic reactive oxygen radicals has been discussed numerous times. The observations that iron induces aggregation of inert alpha-synuclein and beta-amyloid peptides to toxic aggregates have reinforced the critical role of iron in OS-induced pathogenesis of neurodegeneration, supporting the notion that a combination of iron chelation and antioxidant therapy may be one significant approach for neuroprotection. Tea flavonoids (catechins) have been reported to possess divalent metal chelating, antioxidant, and anti-inflammatory activities, to penetrate the brain barrier and to protect neuronal death in a wide array of cellular and animal models of neurological diseases. This review aims to shed light on the multipharmacological neuroprotective activities of green tea catechins with special emphasis on their brain-permeable, nontoxic, transitional metal (iron and copper)-chelatable/radical scavenger properties.
Collapse
|
283
|
Agiostratidou G, Muros RM, Shioi J, Marambaud P, Robakis NK. The cytoplasmic sequence of E-cadherin promotes non-amyloidogenic degradation of A beta precursors. J Neurochem 2006; 96:1182-8. [PMID: 16417575 DOI: 10.1111/j.1471-4159.2005.03616.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The presenilin (PS)/gamma-secretase system promotes production of the A beta (A beta) peptides by mediating cleavage of amyloid precursor protein (APP) at the gamma-sites. This system is also involved in the processing of type-I transmembrane proteins, including APP, cadherins and Notch1 receptors, at the epsilon-cleavage site, resulting in the production of peptides containing the intracellular domains (ICDs) of the cleaved proteins. Emerging evidence shows that these peptides have important biological functions, raising the possibility that their inhibition by gamma-secretase inhibitors may be detrimental to the cell. Here, we show that peptide E-Cad/CTF2, produced by the PS1/gamma-secretase processing of E-cadherin, promotes the lysosomal/endosomal degradation of the transmembrane APP derivatives, C99 and C83, and inhibits production of the APP ICD (AICD). In addition, E-Cad/CTF2 decreases accumulation of total secreted A beta. These data suggest a novel method to promote the non-amyloidogenic degradation of A beta precursors and to inhibit A beta production.
Collapse
|
284
|
Réchards M, Xia W, Oorschot V, van Dijk S, Annaert W, Selkoe DJ, Klumperman J. Presenilin-1-mediated Retention of APP Derivatives in Early Biosynthetic Compartments. Traffic 2006; 7:354-64. [PMID: 16497228 DOI: 10.1111/j.1600-0854.2006.00388.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Processing of the amyloid precursor protein (APP) leads to the production of amyloid-beta (Abeta), the major component of extracellular plaques in the brains of Alzheimer's disease (AD) patients. Presenilin-1 (PS-1) plays a key role in the final step of Abeta formation, the gamma-secretase cleavage. Previously, we showed that PS-1 is retained in pre-Golgi compartments by incorporation into COPI-coated membranes of the vesicular tubular clusters (VTCs) between endoplasmic reticulum (ER) and Golgi complex. Here, we show that PS-1 also mediates the retention of the beta-cleavage-derived APP-C-terminal fragment (CTFbeta) and/or Abeta in pre-Golgi membranes. Overexpression of PS-1 increased the percentage of CTFbeta and/or Abeta in VTCs as well as their distribution to COPI-coated VTC membranes. By contrast, overexpression of the dominant-negative aspartate mutant PS-1(D257A) or PS-knockout decreased incorporation of these APP derivatives into COPI-coated membranes. Sorting of APP derivatives to COPI-coated VTC membranes was not depending on the APP cytosolic tail. In post-Golgi compartments, PS-1 expression enhanced the association of full-length APP/APPs with endosomal compartments at the expense of plasma membrane-bound APP. We conclude that PS-1, in addition to its role in gamma-secretase cleavage, is also required for the subcellular routing of APP and its derivatives. Malfunctioning of PS-1 in this role may have important consequences for the progress of AD.
Collapse
Affiliation(s)
- Marloes Réchards
- Cell Microscopy Center, Department of Cell Biology, University Medical Center and Institute for Biomembranes, 3584 CX Utrecht, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
285
|
Cho HW, Kim JH, Choi S, Kim HJ. Phospholipase A2 is involved in muscarinic receptor-mediated sAPPalpha release independently of cyclooxygenase or lypoxygenase activity in SH-SY5Y cells. Neurosci Lett 2006; 397:214-8. [PMID: 16406345 DOI: 10.1016/j.neulet.2005.12.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Revised: 11/12/2005] [Accepted: 12/07/2005] [Indexed: 12/13/2022]
Abstract
The release of soluble amyloid precursor protein alpha (sAPPalpha), produced during alpha-secretase processing by cleavage within the beta amyloid peptide domain of APP, is highly regulated by several external and internal signals. Because evidence suggests the involvement of inflammatory processes in the pathology of Alzheimer's disease and APP formation, we examined the involvement of the phospholipase A2 (PLA2) pathway and of its downstream cyclooxygenase (COX) and lipoxygenase (LOX) pathways in the regulation of sAPPalpha, release induced by muscarinic receptor activation in SH-SY5Y cells. The amount of sAPP released into the culture medium was analyzed using a monoclonal 6E10 antibody detecting sAPPalpha. Treatment with the PLA2 inhibitor, manoalide, blocked the release of oxoM (muscarinic receptor agonist)-stimulated sAPPalpha, and the muscarinic receptor-mediated sAPPalpha release was increased by the non-selective PLA2 activator mellitin. COX and LOX inhibitors inhibited exogenous AA-induced sAPPalpha release, but upregulated basal constitutive sAPPalpha release. However, treatment with COX or LOX inhibitors failed to significantly change oxoM-stimulated sAPPalpha release, and furthermore, muscarinic receptor activation inhibited AA-stimulated COX activity. Our results suggest that sAPPalpha release induced by muscarinic receptor activation is regulated by AA generation via PLA2 activation independently of COX and LOX activities, but that the COX and LOX pathways are possibly involved in the constitutive release of sAPPalpha.
Collapse
Affiliation(s)
- Hye-Won Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, 11-1, Daehyun-Dong, Seodaemun-Gu, Seoul 120-750, Republic of Korea
| | | | | | | |
Collapse
|
286
|
Costantini C, Weindruch R, Della Valle G, Puglielli L. A TrkA-to-p75NTR molecular switch activates amyloid beta-peptide generation during aging. Biochem J 2006; 391:59-67. [PMID: 15966860 PMCID: PMC1237139 DOI: 10.1042/bj20050700] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aging is the single most important risk factor for AD (Alzheimer's disease). However, the molecular events that connect normal aging to AD are mostly unknown. The abnormal accumulation of Abeta (amyloid beta-peptide) in the form of senile plaques is one of the main characteristics of AD. In the present study, we show that two members of the neurotrophin receptor superfamily, TrkA (tyrosine kinase receptor A) and p75NTR (p75 neurotrophin receptor), differentially regulate the processing of APP (amyloid precursor protein): TrkA reduces, whereas p75NTR activates, beta-cleavage of APP. The p75NTR-dependent effect requires NGF (nerve growth factor) binding and activation of the second messenger ceramide. We also show that normal aging activates Abeta generation in the brain by 'switching' from the TrkA to the p75NTR receptor system. Such an effect is abolished in p75NTR 'knockout' animals, and can be blocked by both caloric restriction and inhibitors of nSMase (neutral sphingomyelinase). In contrast with caloric restriction, which prevents the age-associated up-regulation of p75NTR expression, nSMase inhibitors block the activation of ceramide. When taken together, these results indicate that the p75NTR-ceramide signalling pathway activates the rate of Abeta generation in an age-dependent fashion, and provide a new target for both the understanding and the prevention of late-onset AD.
Collapse
Affiliation(s)
- Claudio Costantini
- *Department of Medicine, University of Wisconsin-Madison, Wm. S. Middleton Memorial Veteran's Hospital, Madison, WI 53705, U.S.A
| | - Richard Weindruch
- *Department of Medicine, University of Wisconsin-Madison, Wm. S. Middleton Memorial Veteran's Hospital, Madison, WI 53705, U.S.A
| | | | - Luigi Puglielli
- *Department of Medicine, University of Wisconsin-Madison, Wm. S. Middleton Memorial Veteran's Hospital, Madison, WI 53705, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
287
|
Endres K, Postina R, Schroeder A, Mueller U, Fahrenholz F. Shedding of the amyloid precursor protein-like protein APLP2 by disintegrin-metalloproteinases. FEBS J 2005; 272:5808-20. [PMID: 16279945 DOI: 10.1111/j.1742-4658.2005.04976.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cleavage of the amyloid precursor protein (APP) within the amyloid-beta (Abeta) sequence by the alpha-secretase prevents the formation of toxic Abeta peptides. It has been shown that the disintegrin-metalloproteinases ADAM10 and TACE (ADAM17) act as alpha-secretases and stimulate the generation of a soluble neuroprotective fragment of APP, APPsalpha. Here we demonstrate that the related APP-like protein 2 (APLP2), which has been shown to be essential for development and survival of mice, is also a substrate for both proteinases. Overexpression of either ADAM10 or TACE in HEK293 cells increased the release of neurotrophic soluble APLP2 severalfold. The strongest inhibition of APLP2 shedding in neuroblastoma cells was observed with an ADAM10-preferring inhibitor. Transgenic mice with neuron-specific overexpression of ADAM10 showed significantly increased levels of soluble APLP2 and its C-terminal fragments. To elucidate a possible regulatory mechanism of APLP2 shedding in the neuronal context, we examined retinoic acid-induced differentiation of neuroblastoma cells. Retinoic acid treatment of two neuroblastoma cell lines upregulated the expression of both APLP2 and ADAM10, thus leading to an increased release of soluble APLP2.
Collapse
Affiliation(s)
- Kristina Endres
- Institute of Biochemistry, Johannes Gutenberg-University Mainz, Germany
| | | | | | | | | |
Collapse
|
288
|
Swanson T, Knittel LM, Coate T, Farley S, Snyder M, Copenhaver P. The insect homologue of the amyloid precursor protein interacts with the heterotrimeric G protein Go alpha in an identified population of migratory neurons. Dev Biol 2005; 288:160-78. [PMID: 16229831 PMCID: PMC2862231 DOI: 10.1016/j.ydbio.2005.09.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2005] [Revised: 09/01/2005] [Accepted: 09/06/2005] [Indexed: 10/25/2022]
Abstract
The amyloid precursor protein (APP) is the source of Abeta fragments implicated in the formation of senile plaques in Alzheimer's disease (AD). APP-related proteins are also expressed at high levels in the embryonic nervous system and may serve a variety of developmental functions, including the regulation of neuronal migration. To investigate this issue, we have cloned an orthologue of APP (msAPPL) from the moth, Manduca sexta, a preparation that permits in vivo manipulations of an identified set of migratory neurons (EP cells) within the developing enteric nervous system. Previously, we found that EP cell migration is regulated by the heterotrimeric G protein Goalpha: when activated by unknown receptors, Goalpha induces the onset of Ca2+ spiking in these neurons, which in turn down-regulates neuronal motility. We have now shown that msAPPL is first expressed by the EP cells shortly before the onset of migration and that this protein undergoes a sequence of trafficking, processing, and glycosylation events that correspond to discrete phases of neuronal migration and differentiation. We also show that msAPPL interacts with Goalpha in the EP cells, suggesting that msAPPL may serve as a novel G-protein-coupled receptor capable of modulating specific aspects of migration via Goalpha-dependent signal transduction.
Collapse
Affiliation(s)
- T.L. Swanson
- Dept of Cell & Developmental Biology, L-215; Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239
| | - L. M. Knittel
- Dept of Cell & Developmental Biology, L-215; Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239
| | - T.M. Coate
- Dept of Cell & Developmental Biology, L-215; Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239
| | - S.M. Farley
- Dept of Cell & Developmental Biology, L-215; Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239
| | - M.A. Snyder
- Dept of Cell & Developmental Biology, L-215; Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239
| | - P.F. Copenhaver
- Dept of Cell & Developmental Biology, L-215; Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239
| |
Collapse
|
289
|
Li X, Bürklen T, Yuan X, Schlattner U, Desiderio DM, Wallimann T, Homayouni R. Stabilization of ubiquitous mitochondrial creatine kinase preprotein by APP family proteins. Mol Cell Neurosci 2005; 31:263-72. [PMID: 16260146 DOI: 10.1016/j.mcn.2005.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2005] [Revised: 08/06/2005] [Accepted: 09/22/2005] [Indexed: 11/20/2022] Open
Abstract
Amyloid precursor protein (APP) is involved in the pathogenesis of Alzheimer's disease (AD). However, the physiological role of APP and its family members is still unclear. To gain insights into APP function, we used a proteomic approach to identify APP interacting proteins. We report here for the first time a direct interaction between the C-terminal region of APP family proteins and ubiquitous mitochondrial creatine kinase (uMtCK). This interaction was confirmed in vitro as well as in cultured cells and in brain. Interestingly, expression of full-length and C-terminal domain of APP family proteins stabilized uMtCK preprotein in cultured cells. Our data suggest that APP may regulate cellular energy levels and mitochondrial function via a direct interaction and stabilization of uMtCK.
Collapse
Affiliation(s)
- Xiaofan Li
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | |
Collapse
|
290
|
Reinhard C, Hébert SS, De Strooper B. The amyloid-beta precursor protein: integrating structure with biological function. EMBO J 2005; 24:3996-4006. [PMID: 16252002 PMCID: PMC1356301 DOI: 10.1038/sj.emboj.7600860] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Accepted: 10/10/2005] [Indexed: 12/28/2022] Open
Abstract
Proteolytic processing of the amyloid-beta precursor protein (APP) generates the Abeta amyloid peptide of Alzheimer's disease. The biological function of APP itself remains, however, unclear. In the current review, we study in detail the different subdomains of APP and try to assign functional significance to particular structures identified in the protein.
Collapse
Affiliation(s)
- Constanze Reinhard
- Laboratory for Neuronal Cell Biology and Gene Transfer, Department of Human Genetics, Center for Human Genetics, KU Leuven and VIB, Leuven, Belgium
| | - Sébastien S Hébert
- Laboratory for Neuronal Cell Biology and Gene Transfer, Department of Human Genetics, Center for Human Genetics, KU Leuven and VIB, Leuven, Belgium
| | - Bart De Strooper
- Laboratory for Neuronal Cell Biology and Gene Transfer, Department of Human Genetics, Center for Human Genetics, KU Leuven and VIB, Leuven, Belgium
- Laboratory for Neuronal Cell Biology and Gene Transfer, Department of Human Genetics, Center for Human Genetics, KU Leuven and VIB, Herestraat 49, Leuven 3000, Belgium. Tel.: +32 16 346227; Fax: +32 16 347181; E-mail:
| |
Collapse
|
291
|
Andersson C, Fernandez-Rodriguez J, Laos S, Baeckström D, Haass C, Hansson G. Shedding and gamma-secretase-mediated intramembrane proteolysis of the mucin-type molecule CD43. Biochem J 2005; 387:377-84. [PMID: 15540986 PMCID: PMC1134965 DOI: 10.1042/bj20041387] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CD43 is a transmembrane molecule that contains a 123-aminoacids-long cytoplasmic tail and a highly O-glycosylated extracellular domain of mucin type. Endogenous CD43 expressed in COLO 205, K562 and Jurkat cells revealed a membrane-associated, 20 kDa CD43-specific cytoplasmic tail fragment (CD43-CTF) upon inhibition of gamma-secretase. This fragment was formed by an extracellular cleavage, as it was not accumulated after treating cells with 1,10-phenanthroline, a metalloprotease inhibitor. When CD43 was transfected into HEK-293 cells expressing dominant-negative PS1 (presenilin-1), the CD43-CTF was accumulated, but not in cells with wild-type PS1. Owing to its accumulation in the presence of a non-functional PS variant, it may thus be a novel gamma-secretase substrate. This CTF is formed by an extracellular cleavage close to the membrane, is a fragment that can be concluded to be a substrate for gamma-secretase. However, the intracellular gamma-secretase product has not been possible to detect, suggesting a quick processing of this product. During normal growth the CTF was not found without gamma-secretase inhibition, but when the cells (COLO 205) were very confluent the fragment could be detected. The intracellular domain of CD43 has previously been shown to contain a functional nuclear localization signal, and has been suggested to be involved in gene activation. From this and the present results, a novel way to explain how mucin-type molecules may transduce intracellular signals can be proposed.
Collapse
Affiliation(s)
- Christian X. Andersson
- *Department of Medical Biochemistry, Göteborg University, Medicinaregatan 9A, 413 90 Gothenburg, Sweden
| | - Julia Fernandez-Rodriguez
- *Department of Medical Biochemistry, Göteborg University, Medicinaregatan 9A, 413 90 Gothenburg, Sweden
| | - Sirle Laos
- *Department of Medical Biochemistry, Göteborg University, Medicinaregatan 9A, 413 90 Gothenburg, Sweden
| | - Dan Baeckström
- *Department of Medical Biochemistry, Göteborg University, Medicinaregatan 9A, 413 90 Gothenburg, Sweden
| | - Christian Haass
- †Adolf-Butenandt-Institute, Department of Biochemistry, Laboratory for Alzheimer's and Parkinson's Disease Research, Schillerstr. 44, Ludwig-Maximilians-University, D-80336 Munich, Germany
| | - Gunnar C. Hansson
- *Department of Medical Biochemistry, Göteborg University, Medicinaregatan 9A, 413 90 Gothenburg, Sweden
- To whom correspondence should be addressed (email )
| |
Collapse
|
292
|
Ryan KA, Pimplikar SW. Activation of GSK-3 and phosphorylation of CRMP2 in transgenic mice expressing APP intracellular domain. ACTA ACUST UNITED AC 2005; 171:327-35. [PMID: 16230462 PMCID: PMC2171208 DOI: 10.1083/jcb.200505078] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Amyloid precursor protein (APP), implicated in Alzheimer's disease, is a trans-membrane protein of undetermined function. APP is cleaved by γ-secretase that releases the APP intracellular domain (AICD) in the cytoplasm. In vitro studies have implicated AICD in cell signaling and transcriptional regulation, but its biologic relevance has been uncertain and its in vivo function has not been examined. To investigate its functional role, we generated AICD transgenic mice, and found that AICD causes significant biologic changes in vivo. AICD transgenic mice show activation of glycogen synthase kinase-3β (GSK-3β) and phosphorylation of CRMP2 protein, a GSK-3β substrate that plays a crucial role in Semaphorin3a-mediated axonal guidance. Our data suggest that AICD is biologically relevant, causes significant alterations in cell signaling, and may play a role in axonal elongation or pathfinding.
Collapse
Affiliation(s)
- Kathleen A Ryan
- Department of Pathology and Cell Biology Program, Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|
293
|
Kretzschmar D. Neurodegenerative mutants in Drosophila: a means to identify genes and mechanisms involved in human diseases? INVERTEBRATE NEUROSCIENCE 2005; 5:97-109. [PMID: 16187075 DOI: 10.1007/s10158-005-0005-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Accepted: 07/20/2005] [Indexed: 01/10/2023]
Abstract
There are 50 ways to leave your lover (Simon 1987) but many more to kill your brain cells. Several neurodegenerative diseases in humans, like Alzheimer's disease, have been intensely studied but the underlying cellular and molecular mechanisms are still unknown for most of them. For those syndromes where associated gene products have been identified their biochemistry and physiological as well as pathogenic function is often still under debate. This is in part due to the inherent limitations of genetic analyses in humans and other mammals and therefore experimentally accessible invertebrate in vivo models, such as Caenorhabditis elegans and Drosophila melanogaster, have recently been introduced to investigate neurodegenerative syndromes. Several laboratories have used transgenic approaches in Drosophila to study the human genes associated with neurodegenerative diseases. This has added substantially to our understanding of the mechanisms leading to neurodegenerative diseases in humans. The isolation and characterization of Drosophila mutants, which display a variety of neurodegenerative phenotypes, also provide valuable insights into genes, pathways, and mechanisms causing neurodegeneration. So far only about two dozen such mutants have been described but already their characterization reveals an involvement of various cellular functions in neurodegeneration, ranging from preventing oxidative stress to RNA editing. Some of the isolated genes can already be associated with human neurodegenerative diseases and hopefully the isolation and characterization of more of these mutants, together with an analysis of homologous genes in vertebrate models, will provide insights into the genetic and molecular basis of human neurodegenerative diseases.
Collapse
Affiliation(s)
- Doris Kretzschmar
- Center for Research on Occupational and Environmental Toxicology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97201, USA.
| |
Collapse
|
294
|
Oh-hashi K, Nagai T, Tanaka T, Yu H, Hirata Y, Kiuchi K. Determination of hypoxic effect on neprilysin activity in human neuroblastoma SH-SY5Y cells using a novel HPLC method. Biochem Biophys Res Commun 2005; 334:380-5. [PMID: 16005431 DOI: 10.1016/j.bbrc.2005.06.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Accepted: 06/20/2005] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is characterized by extracellular deposition of amyloid-beta-peptide (Abeta), which is closely associated with the metabolic balance between Abeta production and clearance activities. Neprilysin is one of the important enzymes to degrade Abeta in the brain and alternation of its activity would contribute to the AD neuropathology. However, measurement of neprilysin activity in neuronal cells, especially the extracellular activity, is very difficult because of its weak activities. In the present study, we established a sensitive method enough to estimate extracellular neprilysin activity of living cell cultivated in a 96-well plate using HPLC-fluorometric system, and investigated the effect of hypoxia, a closely associated event with neurodegenerative diseases, on neprilysin activity of human neuroblastoma SH-SY5Y cells. We demonstrated that chronic but not acute hypoxia significantly attenuated neprilysin activity without any alterations of neprilysin gene expression. The present study suggests that chronic hypoxia may down-regulate extracellular neprilysin activity of neuronal cells to impair Abeta degradation and associate with the development of amyloid pathology.
Collapse
Affiliation(s)
- Kentaro Oh-hashi
- Department of Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | | | | | | | | | | |
Collapse
|
295
|
Andersen OM, Reiche J, Schmidt V, Gotthardt M, Spoelgen R, Behlke J, von Arnim CAF, Breiderhoff T, Jansen P, Wu X, Bales KR, Cappai R, Masters CL, Gliemann J, Mufson EJ, Hyman BT, Paul SM, Nykjaer A, Willnow TE. Neuronal sorting protein-related receptor sorLA/LR11 regulates processing of the amyloid precursor protein. Proc Natl Acad Sci U S A 2005; 102:13461-6. [PMID: 16174740 PMCID: PMC1224625 DOI: 10.1073/pnas.0503689102] [Citation(s) in RCA: 499] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
sorLA (Sorting protein-related receptor) is a type-1 membrane protein of unknown function that is expressed in neurons. Its homology to sorting receptors that shuttle between the plasma membrane, endosomes, and the Golgi suggests a related function in neuronal trafficking processes. Because expression of sorLA is reduced in the brain of patients with Alzheimer's disease (AD), we tested involvement of this receptor in intracellular transport and processing of the amyloid precursor protein (APP) to the amyloid beta-peptide (Abeta), the principal component of senile plaques. We demonstrate that sorLA interacts with APP in vitro and in living cells and that both proteins colocalize in endosomal and Golgi compartments. Overexpression of sorLA in neurons causes redistribution of APP to the Golgi and decreased processing to Abeta, whereas ablation of sorLA expression in knockout mice results in increased levels of Abeta in the brain similar to the situation in AD patients. Thus, sorLA acts as a sorting receptor that protects APP from processing into Abeta and thereby reduces the burden of amyloidogenic peptide formation. Consequently, reduced receptor expression in the human brain may increase Abeta production and plaque formation and promote spontaneous AD.
Collapse
Affiliation(s)
- Olav M Andersen
- Max Delbrueck Center for Molecular Medicine, D-13125 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
296
|
Kalvodova L, Kahya N, Schwille P, Ehehalt R, Verkade P, Drechsel D, Simons K. Lipids as modulators of proteolytic activity of BACE: involvement of cholesterol, glycosphingolipids, and anionic phospholipids in vitro. J Biol Chem 2005; 280:36815-23. [PMID: 16115865 DOI: 10.1074/jbc.m504484200] [Citation(s) in RCA: 219] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The beta-secretase, BACE, is a membrane spanning aspartic protease, which cleaves the amyloid precursor protein (APP) in the first step of proteolytic processing leading to the formation of the neurotoxic beta-amyloid peptide (Abeta). Previous results have suggested that the regulation of beta-secretase and BACE access to APP is lipid dependent, and involves lipid rafts. Using the baculovirus expression system, we have expressed recombinant human full-length BACE in insect cells and purified milligram amounts to homogeneity. We have studied partitioning of fluorophor-conjugated BACE between the liquid ordered and disordered phases in giant (10-150 mum) unilamellar vesicles, and found approximately 20% to associate with the raft-like, liquid-ordered phase; the fraction associated with liquid-ordered phase increased upon cross-linking of raft lipids. To examine involvement of individual lipid species in modulating BACE activity, we have reconstituted the purified BACE in large ( approximately 100 nm) unilamellar vesicles, and determined its specific activity in vesicles of various lipid compositions. We have identified 3 groups of lipids that stimulate proteolytic activity of BACE: 1) neutral glycosphingolipids (cerebrosides), 2) anionic glycerophospholipids, and 3) sterols (cholesterol).
Collapse
Affiliation(s)
- Lucie Kalvodova
- Max-Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
297
|
Carey RM, Balcz BA, Lopez-Coviella I, Slack BE. Inhibition of dynamin-dependent endocytosis increases shedding of the amyloid precursor protein ectodomain and reduces generation of amyloid beta protein. BMC Cell Biol 2005; 6:30. [PMID: 16095541 PMCID: PMC1208872 DOI: 10.1186/1471-2121-6-30] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Accepted: 08/11/2005] [Indexed: 11/13/2022] Open
Abstract
Background The amyloid precursor protein (APP) is transported via the secretory pathway to the cell surface, where it may be cleaved within its ectodomain by α-secretase, or internalized within clathrin-coated vesicles. An alternative proteolytic pathway occurs within the endocytic compartment, where the sequential action of β- and γ-secretases generates the amyloid β protein (Aβ). In this study, we investigated the effects of modulators of endocytosis on APP processing. Results Human embryonic kidney cells were transfected with a dominant negative mutant of dynamin I, an important mediator of clathrin-dependent endocytosis, and APP proteolysis was analyzed. Overexpression of the mutant dynamin (dyn I K44A) resulted in increased shedding of the APP ectodomain (sAPPα), accumulation of the C-terminal α-secretase product C83, and a reduction in the release of Aβ. Levels of mature APP on the cell surface were increased in cells expressing dyn I K44A, and internalization of surface-immunolabeled APP, assessed by fluorescence microscopy, was inhibited. Dynamin is a substrate for protein kinase C (PKC), and it was hypothesized that activators of PKC, which are known to stimulate α-secretase-mediated cleavage of APP, might exert their effects by inhibiting dynamin-dependent endocytosis. However, the internalization of surface-biotinylated APP was unaffected by treatment of cells with phorbol 12-myristate 13-acetate in the presence of the α-secretase inhibitor TAPI-1. Conclusion The results indicate that APP is internalized by a dynamin-dependent process, and suggest that alterations in the activity of proteins that mediate endocytosis might lead to significant changes in Aβ production.
Collapse
Affiliation(s)
- Robyn M Carey
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
| | - Brigitte A Balcz
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
- Gemeinnützige Salzburger Landeskliniken Betriebsgesellschaft mbH, Universitätsklinik für Innere Medizin III, Paracelsus Medizinische Privatuniversität, Müllner Hauptstrasse 48, A-5020 Salzburg, Austria
| | - Ignacio Lopez-Coviella
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
| | - Barbara E Slack
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
| |
Collapse
|
298
|
Leyssen M, Ayaz D, Hébert SS, Reeve S, De Strooper B, Hassan BA. Amyloid precursor protein promotes post-developmental neurite arborization in the Drosophila brain. EMBO J 2005; 24:2944-55. [PMID: 16052209 PMCID: PMC1187942 DOI: 10.1038/sj.emboj.7600757] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Accepted: 07/06/2005] [Indexed: 01/12/2023] Open
Abstract
The mechanisms regulating the outgrowth of neurites during development, as well as after injury, are key to the understanding of the wiring and functioning of the brain under normal and pathological conditions. The amyloid precursor protein (APP) is involved in the pathogenesis of Alzheimer's disease (AD). However, its physiological role in the central nervous system is not known. Many physical interactions between APP and intracellular signalling molecules have been described, but their functional relevance remains unclear. We show here that human APP and Drosophila APP-Like (APPL) can induce postdevelopmental axonal arborization, which depends critically on a conserved motif in the C-terminus and requires interaction with the Abelson (Abl) tyrosine kinase. Brain injury induces APPL upregulation in Drosophila neurons, correlating with increased post-traumatic mortality in appl(d) mutant flies. Finally, we also found interactions between APP and the JNK stress kinase cascade. Our findings suggest a role for APP in axonal outgrowth after traumatic brain injury.
Collapse
Affiliation(s)
- Maarten Leyssen
- Laboratory of Neurogenetics, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
- Neuronal Cell Biology and Gene Transfer Laboratory, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
| | - Derya Ayaz
- Laboratory of Neurogenetics, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
| | - Sébastien S Hébert
- Neuronal Cell Biology and Gene Transfer Laboratory, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
| | - Simon Reeve
- Laboratory of Neurogenetics, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
| | - Bart De Strooper
- Neuronal Cell Biology and Gene Transfer Laboratory, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
| | - Bassem A Hassan
- Laboratory of Neurogenetics, Department of Human Genetics, Flanders Interuniversity Institute for Biotechnology (VIB) and University of Leuven, School of Medicine, Leuven, Belgium
- Laboratory of Neurogenetics, Department of Human Genetics—VIB4, VIB and University of Leuven, School of Medicine, Post Box 602, O&N Building, Rm 06.547, Herestraat 49, 3000 Leuven, Belgium. Tel.: +32 16 346226; Fax: +32 16 346218; E-mail:
| |
Collapse
|
299
|
Collin RWJ, van den Hurk WH, Martens GJM. Biosynthesis and differential processing of two pools of amyloid-β precursor protein in a physiologically inducible neuroendocrine cell. J Neurochem 2005; 94:1015-24. [PMID: 16092943 DOI: 10.1111/j.1471-4159.2005.03243.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The amyloid-beta precursor protein (APP) is linked to Alzheimer's disease through its pathological proteolytic processing in the secretory pathway. Nevertheless, surprisingly little is known about the biosynthesis of endogenous APP. We therefore decided to investigate the intracellular fate of newly synthesized APP in a physiologically inducible neuroendocrine cell, the Xenopus intermediate pituitary melanotrope cell. We found that the level of both APP mRNA and protein was about threefold induced in the activated cells of black-adapted animals. Intriguingly, two pools of APP were found, only one of which was up-regulated. This induced pool became readily N- and subsequently O-glycosylated and was eventually proteolytically processed by an alpha-secretase-like cleavage event resulting in a secreted N-terminal and a cell-associated C-terminal APP fragment. Conversely, only the other (non-induced, non-glycosylated and uncleaved) pool became phosphorylated. Thus, we report on the biosynthesis of APP in a physiological context and illuminate the occurrence of two pools of APP, one of which is linked to neuroendocrine cell activation.
Collapse
Affiliation(s)
- Rob W J Collin
- Department of Molecular Animal Physiology, Nijmegen Center for Molecular Life Sciences and Institute for Neuroscience, Radboud University Nijmegen, Nijmegen, The Netherlands
| | | | | |
Collapse
|
300
|
Abstract
The amyloid precursor protein (APP) was initially detected in cells of the central nervous system where it is considered to be involved in the pathogenesis of Alzheimer's disease. However, APP is also found in peripheral organs with exceptionally strong expression in the mammalian epidermis where it fulfils a variety of distinct biological roles. Full length APP appears to facilitate keratinocyte adhesion due to its ability to interact with the extracellular matrix. The C-terminus of APP also serves as adapter protein for binding the motor protein kinesin thereby mediating the centripetal transport of melanosomes in epidermal melanocytes. By the action of alpha-secretase sAPPalpha, the soluble N-terminal portion of APP, is released. sAPPalpha has been shown to be a potent epidermal growth factor thus stimulating proliferation and migration of keratinocytes as well as the exocytic release of melanin by melanocytes. The release of sAPPalpha can be almost completely blocked by inhibiting alpha-secretase with hydroxamic acid-based zinc metalloproteinase inhibitors. In hyperproliferative keratinocytes from psoriatic skin this inhibition results in normalized growth.
Collapse
Affiliation(s)
- Volker Herzog
- Institute of Cell Biology, University of Bonn, Bonn, Germany.
| | | | | | | |
Collapse
|