1
|
Danielsson A, Kogut MM, Maszota-Zieleniak M, Chopra P, Boons GJ, Samsonov SA. Molecular Dynamics-based descriptors of 3-O-Sulfated Heparan Sulfate as Contributors of Protein Binding Specificity. Comput Biol Chem 2022; 99:107716. [DOI: 10.1016/j.compbiolchem.2022.107716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/03/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
|
2
|
Crivelli SM, Luo Q, Stevens JA, Giovagnoni C, van Kruining D, Bode G, den Hoedt S, Hobo B, Scheithauer AL, Walter J, Mulder MT, Exley C, Mold M, Mielke MM, De Vries HE, Wouters K, van den Hove DLA, Berkes D, Ledesma MD, Verhaagen J, Losen M, Bieberich E, Martinez-Martinez P. CERT L reduces C16 ceramide, amyloid-β levels, and inflammation in a model of Alzheimer's disease. Alzheimers Res Ther 2021; 13:45. [PMID: 33597019 PMCID: PMC7890977 DOI: 10.1186/s13195-021-00780-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 01/31/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Dysregulation of ceramide and sphingomyelin levels have been suggested to contribute to the pathogenesis of Alzheimer's disease (AD). Ceramide transfer proteins (CERTs) are ceramide carriers which are crucial for ceramide and sphingomyelin balance in cells. Extracellular forms of CERTs co-localize with amyloid-β (Aβ) plaques in AD brains. To date, the significance of these observations for the pathophysiology of AD remains uncertain. METHODS A plasmid expressing CERTL, the long isoform of CERTs, was used to study the interaction of CERTL with amyloid precursor protein (APP) by co-immunoprecipitation and immunofluorescence in HEK cells. The recombinant CERTL protein was employed to study interaction of CERTL with amyloid-β (Aβ), Aβ aggregation process in presence of CERTL, and the resulting changes in Aβ toxicity in neuroblastoma cells. CERTL was overexpressed in neurons by adeno-associated virus (AAV) in a mouse model of familial AD (5xFAD). Ten weeks after transduction, animals were challenged with behavior tests for memory, anxiety, and locomotion. At week 12, brains were investigated for sphingolipid levels by mass spectrometry, plaques, and neuroinflammation by immunohistochemistry, gene expression, and/or immunoassay. RESULTS Here, we report that CERTL binds to APP, modifies Aβ aggregation, and reduces Aβ neurotoxicity in vitro. Furthermore, we show that intracortical injection of AAV, mediating the expression of CERTL, decreases levels of ceramide d18:1/16:0 and increases sphingomyelin levels in the brain of male 5xFAD mice. CERTL in vivo over-expression has a mild effect on animal locomotion, decreases Aβ formation, and modulates microglia by decreasing their pro-inflammatory phenotype. CONCLUSION Our results demonstrate a crucial role of CERTL in regulating ceramide levels in the brain, in amyloid plaque formation and neuroinflammation, thereby opening research avenues for therapeutic targets of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Simone M. Crivelli
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY USA
- Veterans Affairs Medical Center, Lexington, KY 40502 USA
| | - Qian Luo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Jo A.A. Stevens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Caterina Giovagnoni
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Daan van Kruining
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Gerard Bode
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Sandra den Hoedt
- Department of Internal Medicine, Laboratory Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Barbara Hobo
- Laboratory for Neuroregeneration, Netherlands institute for Neuroscience, Amsterdam, the Netherlands
| | - Anna-Lena Scheithauer
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Monique T. Mulder
- Department of Internal Medicine, Laboratory Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, UK
| | - Matthew Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, UK
| | - Michelle M. Mielke
- Division of Epidemiology, Department of Health Science Research, and Department of Neurology, Mayo Clinic Rochester, Rochester, MN USA
| | - Helga E. De Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands
| | - Daniel L. A. van den Hove
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Dusan Berkes
- Department of Organic Chemistry, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovak Republic
| | - María Dolores Ledesma
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Madrid, Spain
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands institute for Neuroscience, Amsterdam, the Netherlands
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY USA
- Veterans Affairs Medical Center, Lexington, KY 40502 USA
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
3
|
Srivastava AK, Pittman JM, Zerweck J, Venkata BS, Moore PC, Sachleben JR, Meredith SC. β-Amyloid aggregation and heterogeneous nucleation. Protein Sci 2019; 28:1567-1581. [PMID: 31276610 PMCID: PMC6699094 DOI: 10.1002/pro.3674] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 01/09/2023]
Abstract
In this article, we consider the role of heterogeneous nucleation in β-amyloid aggregation. Heterogeneous nucleation is more common and occurs at lower levels of supersaturation than homogeneous nucleation. The nucleation period is also the stage at which most of the polymorphism of amyloids arises, this being one of the defining features of amyloids. We focus on several well-known heterogeneous nucleators of β-amyloid, including lipid surfaces, especially those enriched in gangliosides and cholesterol, and divalent metal ions. These two broad classes of nucleators affect β-amyloid particularly in light of the amphiphilicity of these peptides: the N-terminal region, which is largely polar and charged, contains the metal binding site, whereas the C-terminal region is aliphatic and is important in lipid binding. Notably, these two classes of nucleators can interact cooperatively, aggregation begetting greater aggregation.
Collapse
Affiliation(s)
- Atul K. Srivastava
- Department of PathologyThe University of ChicagoChicagoIllinois
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIllinois
| | - Jay M. Pittman
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIllinois
| | - Jonathan Zerweck
- Department of PathologyThe University of ChicagoChicagoIllinois
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIllinois
| | - Bharat S. Venkata
- Department of PathologyThe University of ChicagoChicagoIllinois
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIllinois
| | | | | | - Stephen C. Meredith
- Department of PathologyThe University of ChicagoChicagoIllinois
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIllinois
| |
Collapse
|
4
|
Young TR, Wedd AG, Xiao Z. Evaluation of Cu(i) binding to the E2 domain of the amyloid precursor protein - a lesson in quantification of metal binding to proteins via ligand competition. Metallomics 2019; 10:108-119. [PMID: 29215101 DOI: 10.1039/c7mt00291b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The extracellular domain E2 of the amyloid precursor protein (APP) features a His-rich metal-binding site (denoted as the M1 site). In conjunction with surrounding basic residues, the site participates in interactions with components of the extracellular matrix including heparins, a class of negatively charged polysaccharide molecules of varying length. This work studied the chemistry of Cu(i) binding to APP E2 with the probe ligands Bcs, Bca, Fz and Fs. APP E2 forms a stable Cu(i)-mediated ternary complex with each of these anionic ligands. The complex with Bca was selected for isolation and characterization and was demonstrated, by native ESI-MS analysis, to have the stoichiometry E2 : Cu(i) : Bca = 1 : 1 : 1. Formation of these ternary complexes is specific for the APP E2 domain and requires Cu(i) coordination to the M1 site. Mutation of the M1 site was consistent with the His ligands being part of the E2 ligand set. It is likely that interactions between the negatively charged probe ligands and a positively charged patch on the surface of APP E2 are one aspect of the generation of the stable ternary complexes. Their formation prevented meaningful quantification of the affinity of Cu(i) binding to the M1 site with these probe ligands. However, the ternary complexes are disrupted by heparin, allowing reliable determination of a picomolar Cu(i) affinity for the E2/heparin complex with the Fz or Bca probe ligands. This is the first documented example of the formation of stable ternary complexes between a Cu(i) binding protein and a probe ligand. The ready disruption of the complexes by heparin identified clear 'tell-tale' signs for diagnosis of ternary complex formation and allowed a systematic review of conditions and criteria for reliable determination of affinities for metal binding via ligand competition. This study also provides new insights into a potential correlation of APP functions regulated by copper binding and heparin interaction.
Collapse
Affiliation(s)
- Tessa R Young
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | |
Collapse
|
5
|
Young TR, Pukala TL, Cappai R, Wedd AG, Xiao Z. The Human Amyloid Precursor Protein Binds Copper Ions Dominated by a Picomolar-Affinity Site in the Helix-Rich E2 Domain. Biochemistry 2018; 57:4165-4176. [DOI: 10.1021/acs.biochem.8b00572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Tessa R. Young
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tara L. Pukala
- Discipline of Chemistry, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Roberto Cappai
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anthony G. Wedd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Zhiguang Xiao
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
6
|
Oliveira J, Costa M, de Almeida MSC, da Cruz e Silva OA, Henriques AG. Protein Phosphorylation is a Key Mechanism in Alzheimer’s Disease. J Alzheimers Dis 2017; 58:953-978. [DOI: 10.3233/jad-170176] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Joana Oliveira
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Márcio Costa
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| | | | - Odete A.B. da Cruz e Silva
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
7
|
Xu F, Davis J, Hoos M, Van Nostrand WE. Mutation of the Kunitz-type proteinase inhibitor domain in the amyloid β-protein precursor abolishes its anti-thrombotic properties in vivo. Thromb Res 2017; 155:58-64. [PMID: 28499154 DOI: 10.1016/j.thromres.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/14/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Kunitz proteinase inhibitor (KPI) domain-containing forms of the amyloid β-protein precursor (AβPP) inhibit cerebral thrombosis. KPI domain-lacking forms of AβPP are abundant in brain. Regions of AβPP other than the KPI domain may also be involved with regulating cerebral thrombosis. To determine the contribution of the KPI domain to the overall function of AβPP in regulating cerebral thrombosis we generated a reactive center mutant that was devoid of anti-thrombotic activity and studied its anti-thrombotic function in vitro and in vivo. METHODS To determine the extent of KPI function of AβPP in regulating cerebral thrombosis we generated a recombinant reactive center KPIR13I mutant devoid of anti-thrombotic activity. The anti-proteolytic and anti-coagulant properties of wild-type and R13I mutant KPI were investigated in vitro. Cerebral thrombosis of wild-type, AβPP knock out and AβPP/KPIR13I mutant mice was evaluated in experimental models of carotid artery thrombosis and intracerebral hemorrhage. RESULTS Recombinant mutant KPIR13I domain was ineffective in the inhibition of pro-thrombotic proteinases and did not inhibit the clotting of plasma in vitro. AβPP/KPIR13I mutant mice were similarly deficient as AβPP knock out mice in regulating cerebral thrombosis in experimental models of carotid artery thrombosis and intracerebral hemorrhage. CONCLUSIONS We demonstrate that the anti-thrombotic function of AβPP primarily resides in the KPI activity of the protein.
Collapse
Affiliation(s)
- Feng Xu
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States
| | - Judianne Davis
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States
| | - Michael Hoos
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States
| | - William E Van Nostrand
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States.
| |
Collapse
|
8
|
Van Nostrand WE. The influence of the amyloid ß-protein and its precursor in modulating cerebral hemostasis. Biochim Biophys Acta Mol Basis Dis 2015; 1862:1018-26. [PMID: 26519139 DOI: 10.1016/j.bbadis.2015.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
Ischemic and hemorrhagic strokes are a significant cause of brain injury leading to vascular cognitive impairment and dementia (VCID). These deleterious events largely result from disruption of cerebral hemostasis, a well-controlled and delicate balance between thrombotic and fibrinolytic pathways in cerebral blood vessels and surrounding brain tissue. Ischemia and hemorrhage are both commonly associated with cerebrovascular deposition of amyloid ß-protein (Aß). In this regard, Aß directly and indirectly modulates cerebral thrombosis and fibrinolysis. Further, major isoforms of the Aß precursor protein (AßPP) function as a potent inhibitor of pro-thrombotic proteinases. The purpose of this review article is to summarize recent research on how cerebral vascular Aß and AßPP influence cerebral hemostasis. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia, edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- William E Van Nostrand
- Department of Neurosurgery, HSC-T12/086, Stony Brook University, Stony Brook, NY 11794-8122, USA; Department of Medicine, HSC-T12/086, Stony Brook University, Stony Brook, NY 11794-8122, USA.
| |
Collapse
|
9
|
Nguyen KV. The human β-amyloid precursor protein: biomolecular and epigenetic aspects. Biomol Concepts 2015; 6:11-32. [DOI: 10.1515/bmc-2014-0041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/22/2015] [Indexed: 11/15/2022] Open
Abstract
AbstractBeta-amyloid precursor protein (APP) is a membrane-spanning protein with a large extracellular domain and a much smaller intracellular domain. APP plays a central role in Alzheimer’s disease (AD) pathogenesis: APP processing generates β-amyloid (Aβ) peptides, which are deposited as amyloid plaques in the brains of AD individuals; point mutations and duplications of APP are causal for a subset of early-onset familial AD (FAD) (onset age <65 years old). However, these mutations in FAD represent a very small percentage of cases (∼1%). Approximately 99% of AD cases are nonfamilial and late-onset, i.e., sporadic AD (SAD) (onset age >65 years old), and the pathophysiology of this disorder is not yet fully understood. APP is an extremely complex molecule that may be functionally important in its full-length configuration, as well as the source of numerous fragments with varying effects on neural function, yet the normal function of APP remains largely unknown. This article provides an overview of our current understanding of APP, including its structure, expression patterns, proteolytic processing and putative functions. Importantly, and for the first time, my recent data concerning its epigenetic regulation, especially in alternative APP pre-mRNA splicing and in the control of genomic rearrangements of the APP gene, are also reported. These findings may provide new directions for investigating the role of APP in neuropathology associated with a deficiency in the enzyme hypoxanthine-guanine phosphoribosyltransferase (HGprt) found in patients with Lesch-Nyhan syndrome (LNS) and its attenuated variants (LNVs). Also, these findings may be of significance for research in neurodevelopmental and neurodegenerative disorders in which the APP gene is involved in the pathogenesis of diseases such as autism, fragile X syndrome (FXS) and AD, with its diversity and complexity, SAD in particular. Accurate quantification of various APP-mRNA isoforms in brain tissues is needed, and antisense drugs are potential treatments.
Collapse
|
10
|
Silva JV, Yoon S, Domingues S, Guimarães S, Goltsev AV, da Cruz E Silva EF, Mendes JFF, da Cruz E Silva OAB, Fardilha M. Amyloid precursor protein interaction network in human testis: sentinel proteins for male reproduction. BMC Bioinformatics 2015; 16:12. [PMID: 25591988 PMCID: PMC4384327 DOI: 10.1186/s12859-014-0432-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 12/16/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amyloid precursor protein (APP) is widely recognized for playing a central role in Alzheimer's disease pathogenesis. Although APP is expressed in several tissues outside the human central nervous system, the functions of APP and its family members in other tissues are still poorly understood. APP is involved in several biological functions which might be potentially important for male fertility, such as cell adhesion, cell motility, signaling, and apoptosis. Furthermore, APP superfamily members are known to be associated with fertility. Knowledge on the protein networks of APP in human testis and spermatozoa will shed light on the function of APP in the male reproductive system. RESULTS We performed a Yeast Two-Hybrid screen and a database search to study the interaction network of APP in human testis and sperm. To gain insights into the role of APP superfamily members in fertility, the study was extended to APP-like protein 2 (APLP2). We analyzed several topological properties of the APP interaction network and the biological and physiological properties of the proteins in the APP interaction network were also specified by gene ontologyand pathways analyses. We classified significant features related to the human male reproduction for the APP interacting proteins and identified modules of proteins with similar functional roles which may show cooperative behavior for male fertility. CONCLUSIONS The present work provides the first report on the APP interactome in human testis. Our approach allowed the identification of novel interactions and recognition of key APP interacting proteins for male reproduction, particularly in sperm-oocyte interaction.
Collapse
Affiliation(s)
- Joana Vieira Silva
- Laboratory of Signal Transduction, Centre for Cell Biology, Health Sciences Department and Biology Department, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Sooyeon Yoon
- Department of Physics, I3N, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Sara Domingues
- Laboratory of Neurosciences, Centre for Cell Biology, Health Sciences Department and Biology Department, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Sofia Guimarães
- Laboratory of Neurosciences, Centre for Cell Biology, Health Sciences Department and Biology Department, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Alexander V Goltsev
- Department of Physics, I3N, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Edgar Figueiredo da Cruz E Silva
- Laboratory of Signal Transduction, Centre for Cell Biology, Health Sciences Department and Biology Department, University of Aveiro, 3810-193, Aveiro, Portugal.
| | | | - Odete Abreu Beirão da Cruz E Silva
- Laboratory of Neurosciences, Centre for Cell Biology, Health Sciences Department and Biology Department, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Centre for Cell Biology, Health Sciences Department and Biology Department, University of Aveiro, 3810-193, Aveiro, Portugal.
- Centro de Biologia Celular, SACS, Edifício 30, Universidade de Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
11
|
Mayer MC, Kaden D, Schauenburg L, Hancock MA, Voigt P, Roeser D, Barucker C, Than ME, Schaefer M, Multhaup G. Novel zinc-binding site in the E2 domain regulates amyloid precursor-like protein 1 (APLP1) oligomerization. J Biol Chem 2014; 289:19019-30. [PMID: 24855651 DOI: 10.1074/jbc.m114.570382] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The amyloid precursor protein (APP) and the APP-like proteins 1 and 2 (APLP1 and APLP2) are a family of multidomain transmembrane proteins possessing homo- and heterotypic contact sites in their ectodomains. We previously reported that divalent metal ions dictate the conformation of the extracellular APP E2 domain (Dahms, S. O., Könnig, I., Roeser, D., Gührs, K.-H., Mayer, M. C., Kaden, D., Multhaup, G., and Than, M. E. (2012) J. Mol. Biol. 416, 438-452), but unresolved is the nature and functional importance of metal ion binding to APLP1 and APLP2. We found here that zinc ions bound to APP and APLP1 E2 domains and mediated their oligomerization, whereas the APLP2 E2 domain interacted more weakly with zinc possessing a less surface-exposed zinc-binding site, and stayed monomeric. Copper ions bound to E2 domains of all three proteins. Fluorescence resonance energy transfer (FRET) analyses examined the effect of metal ion binding to APP and APLPs in the cellular context in real time. Zinc ions specifically induced APP and APLP1 oligomerization and forced APLP1 into multimeric clusters at the plasma membrane consistent with zinc concentrations in the blood and brain. The observed effects were mediated by a novel zinc-binding site within the APLP1 E2 domain as APLP1 deletion mutants revealed. Based upon its cellular localization and its dominant response to zinc ions, APLP1 is mainly affected by extracellular zinc among the APP family proteins. We conclude that zinc binding and APP/APLP oligomerization are intimately linked, and we propose that this represents a novel mechanism for regulating APP/APLP protein function at the molecular level.
Collapse
Affiliation(s)
- Magnus C Mayer
- From the Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Daniela Kaden
- From the Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Linda Schauenburg
- From the Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Mark A Hancock
- the Department of Pharmacology and Therapeutics, McGill University Montreal, Montreal, Quebec H3G 1Y6, Canada
| | - Philipp Voigt
- the Molekulare Pharmakologie und Zellbiologie, Thielallee 67-73, Neurowissenschaftliches Forschungszentrum, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany
| | - Dirk Roeser
- the Leibniz Institute for Age Research, Protein Crystallography Group, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany, and
| | - Christian Barucker
- the Department of Pharmacology and Therapeutics, McGill University Montreal, Montreal, Quebec H3G 1Y6, Canada
| | - Manuel E Than
- the Leibniz Institute for Age Research, Protein Crystallography Group, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany, and
| | - Michael Schaefer
- the Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Medizinische Fakultät der Universität Leipzig, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Gerhard Multhaup
- From the Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany, the Department of Pharmacology and Therapeutics, McGill University Montreal, Montreal, Quebec H3G 1Y6, Canada,
| |
Collapse
|
12
|
Dawkins E, Small DH. Insights into the physiological function of the β-amyloid precursor protein: beyond Alzheimer's disease. J Neurochem 2014; 129:756-69. [PMID: 24517464 PMCID: PMC4314671 DOI: 10.1111/jnc.12675] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 12/21/2022]
Abstract
The β-amyloid precursor protein (APP) has been extensively studied for its role as the precursor of the β-amyloid protein (Aβ) of Alzheimer's disease. However, the normal function of APP remains largely unknown. This article reviews studies on the structure, expression and post-translational processing of APP, as well as studies on the effects of APP in vitro and in vivo. We conclude that the published data provide strong evidence that APP has a trophic function. APP is likely to be involved in neural stem cell development, neuronal survival, neurite outgrowth and neurorepair. However, the mechanisms by which APP exerts its actions remain to be elucidated. The available evidence suggests that APP interacts both intracellularly and extracellularly to regulate various signal transduction mechanisms. This article reviews studies on the structure, expression and post-translational processing of β-amyloid precursor protein (APP), as well as studies on the effects of APP in vitro and in vivo. We conclude that the published data provide strong evidence that APP has a trophic function. APP is likely to be involved in neural stem cell development, neuronal survival, neurite outgrowth and neurorepair. However, the mechanisms by which APP exerts its actions remain to be elucidated. The available evidence suggests that APP interacts both intracellularly and extracellularly to regulate various signal transduction mechanisms.
Collapse
Affiliation(s)
- Edgar Dawkins
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia
| | | |
Collapse
|
13
|
Needham BE, Ciccotosto GD, Cappai R. Combined deletions of amyloid precursor protein and amyloid precursor-like protein 2 reveal different effects on mouse brain metal homeostasis. Metallomics 2014; 6:598-603. [PMID: 24448592 DOI: 10.1039/c3mt00358b] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alterations to the expression of the Amyloid Precursor Protein (APP) and its paralogue Amyloid Precursor-Like Protein 2 (APLP2) affect metal homeostasis in vitro and in vivo. Analysis of the in vivo effects of the APP and APLP2 knockouts on metal homeostasis has been restricted to APP and APLP2 single knockout mice, and up to12 month old animals. To define the redundancy and inter-relationship between the APP and APLP2 genes as regulators of metal homeostasis, and how this is influenced by aging, we investigated copper, iron, zinc and manganese levels in APP and APLP2 single knockout mice as well as homozygous:hemizygous knockout mice at 3, 12 and 18 plus months of age. These studies identified age and genotype dependent changes in metal levels, and established differences in the relative roles played by APP and APLP2 in modulating metal homeostasis.
Collapse
Affiliation(s)
- B Elise Needham
- Department of Pathology, The University of Melbourne, VIC 3010, Australia.
| | | | | |
Collapse
|
14
|
Analysis of the overall structure of the multi-domain amyloid precursor protein (APP). PLoS One 2013; 8:e81926. [PMID: 24324731 PMCID: PMC3852973 DOI: 10.1371/journal.pone.0081926] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
The amyloid precursor protein (APP) and its processing by the α-, β- and γ-secretases is widely believed to play a central role during the development of Alzheimer´s disease. The three-dimensional structure of the entire protein, its physiologic function and the regulation of its proteolytic processing remain, however, largely unclear to date. To gain a deeper understanding of the structure of APP that underlies all of its functions, we first cloned and recombinantly expressed different constructs in E. coli. Using limited proteolysis followed by mass spectrometry and Edman degradation as well as analytical gel permeation chromatography coupled static light scattering, we experimentally analyzed the structural domain boundaries and determined that the large ectodomain of APP consists of exactly two rigidly folded domains - the E1-domain (Leu18-Ala190) and the E2-domain (Ser295-Asp500). Both, the acidic domain (AcD) connecting E1 and E2 as well as the juxtamembrane region (JMR) connecting E2 to the single transmembrane helix are highly flexible and extended. We identified in-between the E1-domain and the AcD an additional domain of conservation and partial flexibility that we termed extension domain (ED, Glu191-Glu227). Using Bio-layer interferometry, pull-down assays and analytical gel filtration experiments we demonstrated that the E1-domain does not tightly interact with the E2-domain, both in the presence and in the absence of heparin. APP hence forms an extended molecule that is flexibly tethered to the membrane. Its multi-domain architecture enables together with the many known functionalities the concomitant performance of several, independent functions, which might be regulated by cellular, compartment specific pH-changes.
Collapse
|
15
|
Villaflores OB, Chen YJ, Chen CP, Yeh JM, Wu TY. Curcuminoids and resveratrol as anti-Alzheimer agents. Taiwan J Obstet Gynecol 2012; 51:515-25. [DOI: 10.1016/j.tjog.2012.09.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2012] [Indexed: 12/24/2022] Open
|
16
|
Libeu CAP, Descamps O, Zhang Q, John V, Bredesen DE. Altering APP proteolysis: increasing sAPPalpha production by targeting dimerization of the APP ectodomain. PLoS One 2012; 7:e40027. [PMID: 22768208 PMCID: PMC3386930 DOI: 10.1371/journal.pone.0040027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 06/04/2012] [Indexed: 12/13/2022] Open
Abstract
One of the events associated with Alzheimer's disease is the dysregulation of α- versus β-cleavage of the amyloid precursor protein (APP). The product of α-cleavage (sAPPα) has neuroprotective properties, while Aβ1-42 peptide, a product of β-cleavage, is neurotoxic. Dimerization of APP has been shown to influence the relative rate of α- and β- cleavage of APP. Thus finding compounds that interfere with dimerization of the APP ectodomain and increase the α-cleavage of APP could lead to the development of new therapies for Alzheimer's disease. Examining the intrinsic fluorescence of a fragment of the ectodomain of APP, which dimerizes through the E2 and Aβ-cognate domains, revealed significant changes in the fluorescence of the fragment upon binding of Aβ oligomers--which bind to dimers of the ectodomain--and Aβ fragments--which destabilize dimers of the ectodomain. This technique was extended to show that RERMS-containing peptides (APP(695) 328-332), disulfiram, and sulfiram also inhibit dimerization of the ectodomain fragment. This activity was confirmed with small angle x-ray scattering. Analysis of the activity of disulfiram and sulfiram in an AlphaLISA assay indicated that both compounds significantly enhance the production of sAPPα by 7W-CHO and B103 neuroblastoma cells. These observations demonstrate that there is a class of compounds that modulates the conformation of the APP ectodomain and influences the ratio of α- to β-cleavage of APP. These compounds provide a rationale for the development of a new class of therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Clare A Peters Libeu
- Buck Institute for Research on Aging, Novato, California, United States of America.
| | | | | | | | | |
Collapse
|
17
|
Heparin induces harmless fibril formation in amyloidogenic W7FW14F apomyoglobin and amyloid aggregation in wild-type protein in vitro. PLoS One 2011; 6:e22076. [PMID: 21779376 PMCID: PMC3135624 DOI: 10.1371/journal.pone.0022076] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 06/14/2011] [Indexed: 12/02/2022] Open
Abstract
Glycosaminoglycans (GAGs) are frequently associated with amyloid deposits in most amyloid diseases, and there is evidence to support their active role in amyloid fibril formation. The purpose of this study was to obtain structural insight into GAG-protein interactions and to better elucidate the molecular mechanism underlying the effect of GAGs on the amyloid aggregation process and on the related cytotoxicity. To this aim, using Fourier transform infrared and circular diochroism spectroscopy, electron microscopy and thioflavin fluorescence dye we examined the effect of heparin and other GAGs on the fibrillogenesis and cytotoxicity of aggregates formed by the amyloidogenic W7FW14 apomyoglobin mutant. Although this protein is unrelated to human disease, it is a suitable model for in vitro studies because it forms amyloid-like fibrils under physiological conditions of pH and temperature. Heparin strongly stimulated aggregation into amyloid fibrils, thereby abolishing the lag-phase normally detected following the kinetics of the process, and increasing the yield of fibrils. Moreover, the protein aggregates were harmless when assayed for cytotoxicity in vitro. Neutral or positive compounds did not affect the aggregation rate, and the early aggregates were highly cytotoxic. The surprising result that heparin induced amyloid fibril formation in wild-type apomyoglobin and in the partially folded intermediate state of the mutant, i.e., proteins that normally do not show any tendency to aggregate, suggested that the interaction of heparin with apomyoglobin is highly specific because of the presence, in protein turn regions, of consensus sequences consisting of alternating basic and non-basic residues that are capable of binding heparin molecules. Our data suggest that GAGs play a dual role in amyloidosis, namely, they promote beneficial fibril formation, but they also function as pathological chaperones by inducing amyloid aggregation.
Collapse
|
18
|
Structure and biochemical analysis of the heparin-induced E1 dimer of the amyloid precursor protein. Proc Natl Acad Sci U S A 2010; 107:5381-6. [PMID: 20212142 DOI: 10.1073/pnas.0911326107] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The amyloid precursor protein (APP) is the key player in Alzheimer's disease pathology, yet APP and its analogues are also essential for neuronal development and cell homeostasis in mammals. We have determined the crystal structure of the entire N-terminal APP-E1 domain consisting of the growth factor like and the copper binding domains at 2.7-A resolution and show that E1 functions as a rigid functional entity. The two subdomains interact tightly in a pH-dependent manner via an evolutionarily conserved interface area. Two E1 entities dimerize upon their interaction with heparin, requiring 8-12 sugar rings to form the heparin-bridged APP-E1 dimer in an endothermic and pH-dependent process that is characterized by a low micromolar dissociation constant. Limited proteolysis confirms that the heparin-bridged E1 dimers obtained in solution correspond to a dimer contact in our crystal, enabling us to model this heparin-[APP-E1](2) complex. Correspondingly, the APP-based signal transduction, cell-cell- and/or cell-ECM interaction should depend on dimerization induced by heparin, as well as on pH, arguing that APP could fulfill different functions depending on its (sub)cellular localization.
Collapse
|
19
|
|
20
|
Physiological and pathological role of alpha-synuclein in Parkinson's disease through iron mediated oxidative stress; the role of a putative iron-responsive element. Int J Mol Sci 2009; 10:1226-60. [PMID: 19399246 PMCID: PMC2672027 DOI: 10.3390/ijms10031226] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Revised: 03/03/2009] [Accepted: 03/11/2009] [Indexed: 12/16/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common progressive neurodegenerative disorder after Alzheimer’s disease (AD) and represents a large health burden to society. Genetic and oxidative risk factors have been proposed as possible causes, but their relative contribution remains unclear. Dysfunction of alpha-synuclein (α-syn) has been associated with PD due to its increased presence, together with iron, in Lewy bodies. Brain oxidative damage caused by iron may be partly mediated by α-syn oligomerization during PD pathology. Also, α-syn gene dosage can cause familial PD and inhibition of its gene expression by blocking translation via a newly identified Iron Responsive Element-like RNA sequence in its 5’-untranslated region may provide a new PD drug target.
Collapse
|
21
|
Short-Term Effects of β-Amyloid25-35Peptide Aggregates on Transmitter Release in Neuromuscular Synapses. J Neuropathol Exp Neurol 2008; 67:250-9. [DOI: 10.1097/nen.0b013e318165e300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Osterfield M, Egelund R, Young LM, Flanagan JG. Interaction of amyloid precursor protein with contactins and NgCAM in the retinotectal system. Development 2008; 135:1189-99. [PMID: 18272596 DOI: 10.1242/dev.007401] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The amyloid precursor protein (APP) plays a central role in Alzheimer's disease, but its actions in normal development are not well understood. Here, a tagged APP ectodomain was used to identify extracellular binding partners in developing chick brain. Prominent binding sites were seen in the olfactory bulb and on retinal axons growing into the optic tectum. Co-precipitation from these tissues and tandem mass spectrometry led to the identification of two associated proteins: contactin 4 and NgCAM. In vitro binding studies revealed direct interactions among multiple members of the APP and contactin protein families. Levels of the APP processing fragment, CTFalpha, were modulated by both contactin 4 and NgCAM. In the developing retinotectal system, APP, contactin 4 and NgCAM are expressed in the retina and tectum in suitable locations to interact. Functional assays revealed regulatory effects of both APP and contactin 4 on NgCAM-dependent growth of cultured retinal axons, demonstrating specific functional interactions among these proteins. These studies identify novel binding and functional interactions among proteins of the APP, contactin and L1CAM families, with general implications for mechanisms of APP action in neural development and disease.
Collapse
Affiliation(s)
- Miriam Osterfield
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
23
|
Kaden D, Munter LM, Joshi M, Treiber C, Weise C, Bethge T, Voigt P, Schaefer M, Beyermann M, Reif B, Multhaup G. Homophilic interactions of the amyloid precursor protein (APP) ectodomain are regulated by the loop region and affect beta-secretase cleavage of APP. J Biol Chem 2008; 283:7271-9. [PMID: 18182389 DOI: 10.1074/jbc.m708046200] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We found previously by fluorescence resonance energy transfer experiments that amyloid precursor protein (APP) homodimerizes in living cells. APP homodimerization is likely to be mediated by two sites of the ectodomain and a third site within the transmembrane sequence of APP. We have now investigated the role of the N-terminal growth factor-like domain in APP dimerization by NMR, biochemical, and cell biological approaches. Under nonreducing conditions, the N-terminal domain of APP formed SDS-labile and SDS-stable complexes. The presence of SDS was sufficient to convert native APP dimers entirely into monomers. Addition of an excess of a synthetic peptide (APP residues 91-116) containing the disulfide bridge-stabilized loop inhibited cross-linking of pre-existing SDS-labile APP ectodomain dimers. Surface plasmon resonance analysis revealed that this peptide specifically bound to the N-terminal domain of APP and that binding was entirely dependent on the oxidation of the thiol groups. By solution-state NMR we detected small chemical shift changes indicating that the loop peptide interacted with a large protein surface rather than binding to a defined pocket. Finally, we studied the effect of the loop peptide added to the medium of living cells. Whereas the levels of alpha-secretory APP increased, soluble beta-cleaved APP levels decreased. Because Abeta40 and Abeta42 decreased to similar levels as soluble beta-cleaved APP, we conclude either that beta-secretase binding to APP was impaired or that the peptide allosterically affected APP processing. We suggest that APP acquires a loop-mediated homodimeric state that is further stabilized by interactions of hydrophobic residues of neighboring domains.
Collapse
Affiliation(s)
- Daniela Kaden
- Institut für Chemie und Biochemie, Freie Universität Berlin, 14195 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Affiliation(s)
- Michael S Wolfe
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
25
|
Schmitt HP. Profiling the culprit in Alzheimer's disease (AD): bacterial toxic proteins - Will they be significant for the aetio-pathogenesis of AD and the transmissible spongiform encephalopathies? Med Hypotheses 2007; 69:596-609. [PMID: 17337124 DOI: 10.1016/j.mehy.2007.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Accepted: 01/02/2007] [Indexed: 11/25/2022]
Abstract
The aetiology of Alzheimer's disease (AD) and the transmissible spongiform encephalopathies (tSEs) is still elusive. The concept that prion protein (PrP(Sc)) is the aetiological agent (infectious protein) in the tSEs has recently been questioned. In AD, the cause of the aberrant cleavage of the beta-amyloid precursor protein (APP), resulting in the production of amyloidogenic Abeta fragments, has yet remained obscure. Moreover, the amyloid hypothesis of AD has been seriously challenged. In both AD and the tSEs, pathogens of various nature, including bacteria, have been discussed as possible causal factors. However, aetiological considerations have completely neglected microbial products such as the bacterial toxic proteins (BTPs). The present paper is aimed at drawing a "culprit profile" of these toxic molecules that can exert, at low-dosage, neuro-degeneration through various effects. Clearly, BTPs may affect cell-surface receptors including modulatory amine transmitter receptor expression, block neuro-transmitter release, increase intra-cellular Ca(2+) levels, affect intra-cellular signal transduction, change cyto-skeletal processing, alter synaptic transmission, influence APP proteolysis, interact with cell surface proteins like PrP(C) or their GPI anchors, act as chaperones inducing conformational change in proteins (e.g., PrP(C) to PrP(Sc)), alter lipid membrane integrity by affecting phospholipases or forming pores and channels, induce vacuolar (spongiform) change and elicit inflammatory reactions with cytokine production including cytokines that were demonstrated in the AD brain. Like PrP(Sc), BTPs can be heat-stable and acid-resistant. BTPs can meet the key-proteins of AD and tSEs in the lipid-rich domains of the plasma membrane called rafts. Basically, this might enable them to initiate a large variety of unfavourable molecular events, eventually resulting in pathogenetic cascades as in AD and the tSEs. All in all, their profile lends support to the hypothesis that BTPs might represent relevant culprits capable to cue and/or promote neuro-degeneration in both AD and the tSEs.
Collapse
Affiliation(s)
- H Peter Schmitt
- Institute of Pathology, Department for Neuropathology, University of Heidelberg, Germany.
| |
Collapse
|
26
|
Abstract
Alzheimer's disease (AD) is associated with the abnormal aggregation of amyloid-beta (Abeta) protein. Abeta and its precursor protein (APP) interact with metal ions such as zinc, copper and iron. Evidence shows that these metals play a role in the precipitation and cytotoxicity of Abeta. Despite recent advances in AD research, there is a lack of therapeutic agents to hinder the apparent aggregation and toxicity of Abeta. Recent studies show that drugs with metal chelating properties could produce a significant reversal of amyloid-beta plaque deposition in vitro and in vivo. Here we discuss the interaction of Abeta with metals, metal dyshomeostasis in the CNS of patients with AD, and the potential therapeutic effects of metal chelators.
Collapse
|
27
|
Rebelo S, Henriques AG, da Cruz e Silva EF, da Cruz e Silva OAB. Effect of cell density on intracellular levels of the Alzheimer's amyloid precursor protein. J Neurosci Res 2004; 76:406-14. [PMID: 15079870 DOI: 10.1002/jnr.20091] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The precise function of APP (Alzheimer's amyloid precursor protein) remains to be fully elucidated, but various lines of evidence suggest that it may be involved in cell adhesion processes. Because APP is a transmembrane glycoprotein, variations in its expression level may have direct bearing on its putative role in cell adhesion. Our results revealed that although APP levels did not change markedly with increasing cell density (ICD), there was a small but reproducible increase in APP expression at subconfluent conditions. Higher expression APP levels led to corresponding increases in the amount of APP processed and secreted APP (sAPP) released into the cell media. Given that phorbol esters stimulate the non-amyloidogenic pathway at the expense of reducing production of Abeta (the peptide found deposited as neuritic plaques in the brains of patients with Alzheimer's disease), thus providing an interesting therapeutic focus, we tested the effect of the phorbol 12-myristate 13-acetate (PMA) on APP processing at ICD. PMA not only stimulated sAPP release at all densities tested, but also produced a corresponding decrease in the intracellular levels of APP. Further experimentation revealed that increased APP expression with ICD was dependent on factors present in conditioned medium. Interestingly, exposing cells to the Abeta peptide itself could mimic these results, thus providing evidence for a potential positive feedback mechanism between Abeta production and intracellular APP levels.
Collapse
Affiliation(s)
- Sandra Rebelo
- Centro de Biologia Celular, Universidade de Aveiro, Aveiro, Portugal
| | | | | | | |
Collapse
|
28
|
Multhaup G, Scheuermann S, Schlicksupp A, Simons A, Strauss M, Kemmling A, Oehler C, Cappai R, Pipkorn R, Bayer TA. Possible mechanisms of APP-mediated oxidative stress in Alzheimer's disease. Free Radic Biol Med 2002; 33:45-51. [PMID: 12086681 DOI: 10.1016/s0891-5849(02)00806-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oxidative stress was presented to play an important role in the pathogenesis of Alzheimer's disease (AD), especially in the early evolution of AD amyloidogenesis and not only as a consequence thereof. The effect of oxidative stress catalysed by transition metals appears to have a critical relevance in AD. Metal-ion homeostasis is severely dysregulated in AD and it was found that experimentally induced disturbances in the homeostasis of Zn(II) and Cu(II) affect the amyloid precursor protein (APP) metabolism. APP itself binds Zn(II) and Cu(II) at nanomolar concentrations and an altered APP metabolism or expression level is believed to result in neurotoxic processes.
Collapse
Affiliation(s)
- Gerd Multhaup
- ZMBH--Center for Molecular Biology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Scheuermann S, Hambsch B, Hesse L, Stumm J, Schmidt C, Beher D, Bayer TA, Beyreuther K, Multhaup G. Homodimerization of amyloid precursor protein and its implication in the amyloidogenic pathway of Alzheimer's disease. J Biol Chem 2001; 276:33923-9. [PMID: 11438549 DOI: 10.1074/jbc.m105410200] [Citation(s) in RCA: 181] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We reported previously that the carbohydrate domain of the amyloid precursor protein is involved in amyloid precursor protein (APP)-APP interactions. Functional in vitro studies suggested that this interaction occurs through the collagen binding site of APP. The physiological significance remained unknown, because it is not understood whether and how APP dimerization occurs in vivo. Here we report that cellular APP exists as homodimers matching best with a two-site model. Consistent with our published crystallographic data, we show that a deletion of the entire sequence after the kunitz protease inhibitor domain did not abolish APP homodimerization, suggesting that two domains are critically involved but that neither is essential for homodimerization. Finally, we generated stabilized dimers by expressing mutant APP with a single cysteine in the ectodomain juxtamembrane region. Mutation of Lys(624) to cysteine produced approximately 6-8-fold more A beta than cells expressing normal APP. Our results suggest that amyloid A beta production can in principle be positively regulated by dimerization in vivo. We suggest that dimerization could be a physiologically important mechanism for regulating the proposed signal activity of APP.
Collapse
Affiliation(s)
- S Scheuermann
- ZMBH, Center for Molecular Biology, University of Heidelberg, Im Neuenheimer Feld 282, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Strausak D, Mercer JF, Dieter HH, Stremmel W, Multhaup G. Copper in disorders with neurological symptoms: Alzheimer's, Menkes, and Wilson diseases. Brain Res Bull 2001; 55:175-85. [PMID: 11470313 DOI: 10.1016/s0361-9230(01)00454-3] [Citation(s) in RCA: 313] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Copper is an essential element for the activity of a number of physiologically important enzymes. Enzyme-related malfunctions may contribute to severe neurological symptoms and neurological diseases: copper is a component of cytochrome c oxidase, which catalyzes the reduction of oxygen to water, the essential step in cellular respiration. Copper is a cofactor of Cu/Zn-superoxide-dismutase which plays a key role in the cellular response to oxidative stress by scavenging reactive oxygen species. Furthermore, copper is a constituent of dopamine-beta-hydroxylase, a critical enzyme in the catecholamine biosynthetic pathway. A detailed exploration of the biological importance and functional properties of proteins associated with neurological symptoms will have an important impact on understanding disease mechanisms and may accelerate development and testing of new therapeutic approaches. Copper binding proteins play important roles in the establishment and maintenance of metal-ion homeostasis, in deficiency disorders with neurological symptoms (Menkes disease, Wilson disease) and in neurodegenerative diseases (Alzheimer's disease). The Menkes and Wilson proteins have been characterized as copper transporters and the amyloid precursor protein (APP) of Alzheimer's disease has been proposed to work as a Cu(II) and/or Zn(II) transporter. Experimental, clinical and epidemiological observations in neurodegenerative disorders like Alzheimer's disease and in the genetically inherited copper-dependent disorders Menkes and Wilson disease are summarized. This could provide a rationale for a link between severely dysregulated metal-ion homeostasis and the selective neuronal pathology.
Collapse
Affiliation(s)
- D Strausak
- Center for Cellular and Molecular Biology, School of Biological and Chemical Sciences, Deakin University, Burwood, Victoria, Australia
| | | | | | | | | |
Collapse
|
31
|
Walter J, Schindzielorz A, Hartung B, Haass C. Phosphorylation of the beta-amyloid precursor protein at the cell surface by ectocasein kinases 1 and 2. J Biol Chem 2000; 275:23523-9. [PMID: 10806211 DOI: 10.1074/jbc.m002850200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The beta-amyloid precursor protein (betaAPP) is one of the rare proteins known to be phosphorylated within its ectodomain. We have shown previously that betaAPP can be phosphorylated within secretory vesicles and at the cell surface (Walter, J., Capell, A., Hung, A. Y. , Langen, H., Schnölzer, M., Thinakaran, G., Sisodia, S. S., Selkoe, D. J., and Haass, C. (1997) J. Biol. Chem. 272, 1896-1903). We have now specifically characterized the phosphorylation of cell surface-located betaAPP and identified two ectoprotein kinases that phosphorylate betaAPP at the outer face of the plasma membrane. By using selective protein kinase inhibitors and by investigating the usage of ATP and GTP as cosubstrates, we demonstrate that membrane-bound betaAPP as well as secreted forms of betaAPP can be phosphorylated by casein kinase (CK) 1- and CK2-like ectoprotein kinases. The ectodomain of betaAPP was also phosphorylated by purified CK1 and CK2 in vitro, but not by protein kinases A and C. Phosphorylation of betaAPP by ectoprotein kinases and by purified CK1 and CK2 occurred within an acidic domain in the N-terminal half of the protein. Heparin strongly inhibited the phosphorylation of cell-surface betaAPP by ecto-CK1 and ecto-CK2, indicating a regulatory role of this extracellular matrix component in betaAPP phosphorylation.
Collapse
Affiliation(s)
- J Walter
- Adolf-Butenandt-Institut, Department of Biochemistry, Laboratory for Alzheimer's Disease Research, Ludwig-Maximilians-Universät München, Schillerstrasse 44, D-80336 Munich, Germany.
| | | | | | | |
Collapse
|
32
|
Abstract
Current assumptions and conclusions in several active areas of amyloid research are examined to see how consistent the data from chosen in vitro and in vivo model systems are with clinical and anatomic observations. These areas include the assembly of amyloid-like fibrils in vitro, the nucleation phenomenon, amyloid fibril structure in vivo and in vitro, common structural components of the amyloids, and the regression of tissue amyloid and proteolysis of amyloid proteins. Divergences and congruencies are highlighted, which in turn suggests caution in the interpretation of present data, greater collaboration and communication among investigators, and, additional areas and techniques for investigation.
Collapse
Affiliation(s)
- R Kisilevsky
- Department of Pathology, Queen's University, The Syl and Molly Apps Research Center, Kingston General Hospital, Ontario, Kingston, K7L 3N6, Canada
| |
Collapse
|
33
|
Melchor JP, Van Nostrand WE. Fibrillar amyloid beta-protein mediates the pathologic accumulation of its secreted precursor in human cerebrovascular smooth muscle cells. J Biol Chem 2000; 275:9782-91. [PMID: 10734132 DOI: 10.1074/jbc.275.13.9782] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cerebrovascular deposition of the amyloid beta-protein (Abeta) is a key pathologic lesion seen in patients with Alzheimer's disease and certain related disorders, including hereditary cerebral hemorrhage with amyloidosis of the Dutch type (HCHWA-D). The deposition of Abeta has pronounced deleterious effects on smooth muscle cells within the cerebral vessel wall. We have previously shown that Abeta(1-40) possessing the E22Q HCHWA-D mutation extensively assembles into fibrils on the surface of cultured human cerebrovascular smooth muscle (HCSM) cells. This cell-surface Abeta fibril formation induces a series of pathologic responses in cultured HCSM cells, including a marked increase in the levels of cell-associated amyloid beta-protein precursor (AbetaPP) and cell death. In the present study, we investigated the relationship between HCSM cell-surface Abeta fibril formation and the striking increase in cell-associated AbetaPP. Time course studies showed that cell-surface HCHWA-D Abeta(1-40) fibril formation occurred rapidly, whereas both the increase in cell-associated AbetaPP and loss of cell viability were delayed responses. Domain analysis using site-specific antibodies indicated that the vast majority of the increase in cell-associated AbetaPP was secreted AbetaPP (sAbetaPP). Localization studies showed that the sAbetaPP was present on the HCSM cell surface. This result raised the possibility that sAbetaPP may bind back to HCSM cell-surface fibrils formed by HCHWA-D Abeta(1-40). Indeed, binding of biotinylated sAbetaPP to fibrillar HCHWA-D Abeta(1-40) was demonstrated by transmission electron microscopy. Furthermore, solid-phase binding assays showed that biotinylated sAbetaPP exhibited dose-dependent, saturable binding to fibrillar (but not soluble) HCHWA-D Abeta(1-40) with k(d) approximately 28 nM. Exon deletion experiments further defined a fragment of sAbetaPP (AbetaPP(18-119)), encoded by AbetaPP exons 2 and 3, to contain the fibrillar Abeta-binding domain. In addition, AbetaPP(18-119) effectively blocked the cell-surface accumulation of sAbetaPP and subsequent cell death in HCSM cells treated with pathogenic Abeta. Together, these findings could explain the accumulation of AbetaPP in cerebrovascular Abeta deposits observed both in vitro and in vivo and may contribute to the pathologic responses evoked by pathogenic forms of Abeta in HCSM cells.
Collapse
Affiliation(s)
- J P Melchor
- Departments of Medicine and Pathology, Health Sciences Center, State University of New York, Stony Brook, New York 11794-8153, USA
| | | |
Collapse
|
34
|
Brückner G, Hausen D, Härtig W, Drlicek M, Arendt T, Brauer K. Cortical areas abundant in extracellular matrix chondroitin sulphate proteoglycans are less affected by cytoskeletal changes in Alzheimer's disease. Neuroscience 1999; 92:791-805. [PMID: 10426522 DOI: 10.1016/s0306-4522(99)00071-8] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the human brain, the distribution of perineuronal nets occurring as lattice-like neuronal coatings of extracellular matrix proteoglycans ensheathing several types of non-pyramidal neurons and subpopulations of pyramidal cells in the cerebral cortex is largely unknown. Since proteoglycans are presumably involved in the pathogenesis of Alzheimer's disease, we analysed the distribution pattern of extracellular chondroitin sulphate proteoglycans in cortical areas, including primary motor, primary auditory and several prefrontal and temporal association areas, in normal human brains and in those showing neuropathological criteria of Alzheimer's disease. In both groups, neurons with perineuronal nets were most numerous in the primary motor cortex (approximately 10% in Brodmann's area 4) and in the primary auditory cortex as a representative of the primary sensory areas. Their number was lower in secondary and higher order association areas. Net-associated pyramidal cells occurred predominantly in layers III and V in motor areas, as well as throughout lower parts of layer III in the primary auditory cortex and neocortical association areas. In the entorhinal cortex, net-associated pyramidal cells were extremely rare. In brains showing hallmarks of Alzheimer's disease, the characteristic patterns of hyperphosphorylated tau protein, stained with the AT8 antibody, largely excluded the zones abundant in perineuronal nets and neuropil-associated chondroitin sulphate proteoglycans. As shown in double-stained sections, pyramidal and non-pyramidal neurons ensheathed by perineuronal nets were virtually unaffected by the formation of neurofibrillary tangles even in severely damaged regions. The distribution patterns of amyloid B deposits overlapped but showed no congruence with that of the extracellular chondroitin sulphate proteoglycans. It can be concluded that low susceptibility of neurons and cortical areas to neurofibrillary changes corresponds with high proportions of aggregating chondroitin sulphate proteoglycans in the neuronal microenvironment.
Collapse
Affiliation(s)
- G Brückner
- Paul Flechsig Institute for Brain Research, Department of Neurochemistry, University of Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Jung SS, Gauthier S, Cashman NR. Beta-amyloid precursor protein is detectable on monocytes and is increased in Alzheimer's disease. Neurobiol Aging 1999; 20:249-57. [PMID: 10588572 DOI: 10.1016/s0197-4580(99)00051-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Using the anti-beta-amyloid precursor protein (betaAPP) monoclonal antibodies 4G8, 6E10 and 22C11 and flow cytometry, we report that human circulating peripheral blood monocytes display surface immunoreactivity for betaAPP. In contrast, circulating lymphocytes do not possess cell surface betaAPP immunoreactivity, despite similar levels of betaAPP expression. Immunoblotting analysis showed that monocytes, but not lymphocytes, possess an 82 kDa C-terminal betaAPP fragment consistent with a processed transmembrane species. Monocyte surface betaAPP was upregulated approximately threefold by activation with lipopolysaccharide and interferon-gamma, activation did not produce detectable betaAPP on the cell surface of lymphocytes. Surface betaAPP immunoreactivity was reduced in a normal aged population compared to normal young controls (Young = 81.07 +/- 13.67 mean fluorescence units, Aged = 36.74 +/- 3.81, p < 0.01), but was significantly increased in AD subjects compared to age-matched healthy controls (AD = 60.31 +/- 7.42, p < 0.05). Our data suggest that a proportion of peripheral A beta may be derived from monocyte/macrophages, and that defects in brain cell processing of betaAPP in AD may be shared by this readily accessible peripheral cell.
Collapse
Affiliation(s)
- S S Jung
- Department of Microbiology & Immunology, Montréal Neurological Institute, McGill University, PQ, Canada
| | | | | |
Collapse
|
36
|
Storey E, Cappai R. The amyloid precursor protein of Alzheimer's disease and the Abeta peptide. Neuropathol Appl Neurobiol 1999; 25:81-97. [PMID: 10215996 DOI: 10.1046/j.1365-2990.1999.00164.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Alzheimer's disease is characterized by the accumulation of beta amyloid peptides in plaques and vessel walls and by the intraneuronal accumulation of paired helical filaments composed of hyperphosphorylated tau. In this review, we concentrate on the biology of amyloid precursor protein, and on the central role of amyloid in the pathogenesis of Alzheimer's disease. Amyloid precursor protein (APP) is part of a super-family of transmembrane and secreted proteins. It appears to have a number of roles, including regulation of haemostasis and mediation of neuroprotection. APP also has potentially important metal and heparin-binding properties, and the current challenge is to synthesize all these varied activities into a coherent view of its function. Cleavage of amyloid precursor protein by beta-and gamma-secretases results in the generation of the Abeta (betaA4) peptide, whereas alpha-secretase cleaves within the Abeta sequence and prevents formation from APP. Recent findings indicate that the site of gamma-secretase cleavage is critical to the development of amyloid deposits; Abeta1-42 is much more amyloidogenic than Abeta1-40. Abeta1-42 formation is favoured by mutations in the two presenilin genes (PS1 and PS2), and by the commonest amyloid precursor protein mutations. Transgenic mouse models of Alzheimer's disease incorporating various mutations in the presenilin gene now exist, and have shown amyloid accumulation and cognitive impairment. Neurofibrillary tangles have not been reproduced in these models, however. While aggregated Abeta is neurotoxic, perhaps via an oxidative mechanism, the relationship between such toxicity and neurofibrillary tangle formation remains a subject of ongoing research.
Collapse
Affiliation(s)
- E Storey
- Van Cleef/Roet Centre for Nervous Diseases, Monash University (Alfred Hospital Campus), Prahran, Victoria, Australia
| | | |
Collapse
|
37
|
Multhaup G, Hesse L, Borchardt T, Ruppert T, Cappai R, Masters CL, Beyreuther K. Autoxidation of amyloid precursor protein and formation of reactive oxygen species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 448:183-92. [PMID: 10079826 DOI: 10.1007/978-1-4615-4859-1_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- G Multhaup
- ZMBH-Center for Molecular Biology Heidelberg, University of Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
38
|
Henry A, Li QX, Galatis D, Hesse L, Multhaup G, Beyreuther K, Masters CL, Cappai R. Inhibition of platelet activation by the Alzheimer's disease amyloid precursor protein. Br J Haematol 1998; 103:402-15. [PMID: 9827912 DOI: 10.1046/j.1365-2141.1998.01005.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The amyloid precursor protein (APP) of Alzheimer's disease is abundantly expressed in the platelet alpha-granule where its role remains unclear. This study describes a novel function for APP in regulating human platelet activation. Preincubation of platelet-rich plasma with recombinant secreted APP (sAPP) isoforms dose-dependently inhibited platelet aggregation and secretion induced by ADP or adrenaline. Similarly, sAPP potently inhibited low-dose thrombin-induced activation in washed platelet suspensions, indicating that the activity does not require plasma cofactors. There were no functional differences between sAPP forms with or without the Kunitz protease inhibitor domain or derived from either alpha- or beta-secretase cleavage. In fact, the N-terminal cysteine-rich region of APP (residues 18-194) was as effective as the entire sAPP region in the inhibition of platelet activation. The inhibitory activity of sAPP correlated with a significant reduction in the agonist-induced production of the arachidonic acid (AA) metabolites thromboxane B2 and prostaglandin E2. However, sAPP did not affect AA-induced platelet aggregation or secretion, indicating the enzymatic conversion of AA was not inhibited. The addition of a threshold dose of AA reversed the sAPP-inhibition of agonist-induced platelet activation. This suggests that sAPP decreases the availability of free AA, although the mechanism is not yet known. These data provide evidence that the release of sAPP upon platelet degranulation may result in negative feedback regulation during platelet activation.
Collapse
Affiliation(s)
- A Henry
- Department of Pathology, University of Melbourne and Mental Health Research Institute of Victoria, Parkville, Australia
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Mulder M, Terwel D. Possible link between lipid metabolism and cerebral amyloid angiopathy in Alzheimer's disease: A role for high-density lipoproteins? HAEMOSTASIS 1998; 28:174-94. [PMID: 10420065 DOI: 10.1159/000022429] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although apolipoprotein E4 (ApoE4) is a well-established risk factor for the development of Alzheimer's disease (AD), it is unclear how ApoE affects the progression of the disease. beta-amyloid (Abeta) is a major constituent of cerebrovascular amyloid deposits in brains of subjects with Alzheimer's disease. In cerebrospinal fluid and in plasma, Abeta is normally present in association with high density lipoproteins (HDL). These lipoproteins may play a role in the removal of excess cholesterol from the brain through interaction with ApoE and heparan sulphate proteoglycans (HSPG) in the subendothelial space of cerebral microvessels. At the same time, HDL may have a role in maintaining Abeta soluble and in mediating its clearance. Therefore, similar factors, e.g. HDL, ApoE and HSPG, may be involved in the regulation of reverse cholesterol transport in the brain and in the processing of Abeta. Alterations in the process of cholesterol secretion from the brain may contribute to the deposition of Abeta in the vascular wall.
Collapse
Affiliation(s)
- M Mulder
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands.
| | | |
Collapse
|
40
|
Breen KC, Coughlan CM, Hayes FD. The role of glycoproteins in neural development function, and disease. Mol Neurobiol 1998; 16:163-220. [PMID: 9588627 DOI: 10.1007/bf02740643] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glycoproteins play key roles in the development, structuring, and subsequent functioning of the nervous system. However, the complex glycosylation process is a critical component in the biosynthesis of CNS glycoproteins that may be susceptible to the actions of toxicological agents or may be altered by genetic defects. This review will provide an outline of the complexity of this glycosylation process and of some of the key neural glycoproteins that play particular roles in neural development and in synaptic plasticity in the mature CNS. Finally, the potential of glycoproteins as targets for CNS disorders will be discussed.
Collapse
Affiliation(s)
- K C Breen
- Neurosciences Institute, Department of Pharmacology and Clinical Pharmacology, University of Dundee, Ninewells Hospital Medical School, Scotland, UK
| | | | | |
Collapse
|
41
|
Beauchemin D, Kisilevsky R. A method based on ICP-MS for the analysis of Alzheimer's amyloid plaques. Anal Chem 1998; 70:1026-9. [PMID: 9511476 DOI: 10.1021/ac970783f] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inductively coupled plasma mass spectrometry (ICP-MS) was combined with flow injection (FI) and a selective extraction procedure (which isolates Alzheimer's amyloid plaques) for the multielemental analysis of plaque cores. FI-ICP-MS was also used to analyze the various reagents involved in the sample preparation to determine whether they were the source of the various elements detected in the plaque samples. An external calibration with matrix-matched standards (in terms of salt concentration) was carried out in all cases. The concentrations of Cr, Mn, Ni, Cu, and Pb were in the 0.2-0.8 mg L-1 range whereas that of Al, Fe, and Zn were 2-20 mg L-1 in the plaque sample. These values can be translated into a microgram per gram level in the plaque core by multiplying them by 29-500 (the exact factor depends on the weight of the plaques, which were not dried to prevent the loss of volatile elements). Although spectroscopic interferences arising from matrix elements of the sample cannot be ruled out in the case of Al, Cr, Ni, Cu, and Fe, the large levels detected (especially for Al and Fe), compared to the much lower or undetectable levels in the various reagents, strongly suggest the accumulation of these elements by the AD patient during life rather than contamination during sample processing.
Collapse
Affiliation(s)
- D Beauchemin
- Department of Chemistry, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
42
|
Kisilevsky R, Fraser PE. A beta amyloidogenesis: unique, or variation on a systemic theme? Crit Rev Biochem Mol Biol 1998; 32:361-404. [PMID: 9383610 DOI: 10.3109/10409239709082674] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
For more than a century amyloid was considered to be an interesting, unique, but inconsequential pathologic entity that rarely caused significant clinical problems. We now recognize that amyloid is not one entity. In vivo it is a uniform organization of a disease, or process, specific protein co-deposited with a set of common structural components. Amyloid has been implicated in the pathogenesis of diseases affecting millions of patients. These range from Alzheimer's disease, adult-onset diabetes, consequences of prolonged renal dialysis, to the historically recognized systemic forms associated with inflammation and plasma cell disturbances. Strong evidence is emerging that even when deposited in local organ sites significant physiologic effects may ensue. With emphasis on A beta amyloid, we review the present definition, classification, and general in vivo pathogenetic events believed to be involved in the deposition of amyloids. This encompasses the need for an adequate amyloid precursor protein pool, whether precursor proteolysis is required prior to deposition, amyloidogenic amino acid sequences, fibrillogenic nucleating particles, and an in vivo microenvironment conducive to fibrillogenesis. The latter includes several components that seem to be part of all amyloids. The role these common components may play in amyloid accumulation, why amyloids tend to be associated with basement membranes, and how one may use these findings for anti-amyloid therapeutic strategies is also examined.
Collapse
Affiliation(s)
- R Kisilevsky
- Department of Pathology, Queen's University, Kingston, Ontario Canada
| | | |
Collapse
|
43
|
Mok SS, Sberna G, Heffernan D, Cappai R, Galatis D, Clarris HJ, Sawyer WH, Beyreuther K, Masters CL, Small DH. Expression and analysis of heparin-binding regions of the amyloid precursor protein of Alzheimer's disease. FEBS Lett 1997; 415:303-7. [PMID: 9357988 DOI: 10.1016/s0014-5793(97)01146-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Deletion mutagenesis studies have suggested that there are two domains within APP which bind heparan sulphate. These domains have been cloned and expressed in the yeast Pichia pastoris. Both recombinant proteins bound to heparin. One domain (APP316-447) was further characterised by binding studies with peptides encompassing this region. Peptides homologous to APP316-346 and APP416-447 were found to bind heparin. Circular dichroism studies show that APP416-447 shifted towards an alpha-helical conformation in the presence of heparin. This study suggests that heparin-binding domains may lie within regions high in alpha-helical structure.
Collapse
Affiliation(s)
- S S Mok
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Multhaup G, Ruppert T, Schlicksupp A, Hesse L, Beher D, Masters CL, Beyreuther K. Reactive oxygen species and Alzheimer's disease. Biochem Pharmacol 1997; 54:533-9. [PMID: 9337068 DOI: 10.1016/s0006-2952(97)00062-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although a consensus that Alzheimer's disease (AD) is a single disease has not been reached yet, the involvement of the amyloid precursor protein (APP) and betaA4 (A beta) in the pathologic changes advances our understanding of the underlying molecular alterations. Increasing evidence implicates oxidative stress in the neurodegenerative process of AD. This hypothesis is based on the toxicity of betaA4 in cell cultures, and the findings that aggregation of betaA4 can be induced by metal-catalyzed oxidation and that free oxygen radicals may be involved in APP metabolism. Another neurological disorder, familial amyotrophic lateral sclerosis (FALS), supports our view that AD and FALS may be linked through a common mechanism. In FALS, SOD-Cu(I) complexes are affected by hydrogen peroxide and free radicals are produced. In AD, the reduction of Cu(II) to Cu(I) by APP involves an electron-transfer reaction and could also lead to a production of hydroxyl radicals. Thus, copper-mediated toxicity of APP-Cu(II)/(I) complexes may contribute to neurodegeneration in AD.
Collapse
Affiliation(s)
- G Multhaup
- ZMBH-Center for Molecular Biology Heidelberg, University of Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
45
|
Gillian AM, McFarlane I, Lucy FM, Overly C, McConlogue L, Breen KC. Individual isoforms of the amyloid beta precursor protein demonstrate differential adhesive potentials to constituents of the extracellular matrix. J Neurosci Res 1997; 49:154-60. [PMID: 9272638 DOI: 10.1002/(sici)1097-4547(19970715)49:2<154::aid-jnr4>3.0.co;2-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The amyloid beta precursor protein (AbetaPP) can exist as a membrane-bound glycoprotein which modulates neural cell adhesion. The adhesion of clones of the AtT20 mouse pituitary cell line, transfected with cDNA coding for the 695 (AbetaPP695) and 751 (AbetaPP751) amino acid forms of the protein, to individual components of the extracellular matrix was determined using a centrifugal shear assay. On laminin, poly-L-lysine, fibronectin, and uncoated glass substrata, the cells transfected with AbetaPP695 (6A1 cells) demonstrated a 50% increase in adhesivity over nontransfected cells, while those transfected with AbetaPP751 (7A1 cells) showed a significant decrease in adhesion. There was, however, a significant increase in the adhesive strength of the 7A1 cells to collagen type IV with no change in the adhesivity of the 6A1 cells when compared with control. These changes in adhesivity could be attributed to changes in the levels of the membrane-bound protein and were not due to the interaction of soluble AbetaPP with elements of the extracellular matrix. These studies provide evidence for differential adhesivities of the constituent AbetaPP isoforms and the possible role of the Kunitz protease inhibitor (KPI) domain in influencing the adhesive properties of the protein backbone.
Collapse
Affiliation(s)
- A M Gillian
- Neurosciences Institute, Department of Pharmacology and Clinical Pharmacology, University of Dundee, Ninewells Hospital Medical School, Scotland
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Although a consensus that Alzheimer's disease (AD) is a single disease has not yet been reached, the involvement of the amyloid precursor protein (APP) and beta A4 (A beta) in the pathologic changes advances our understanding of the underlying molecular alterations. Increasing evidence implicates oxidative stress in the neurodegenerative process of AD. This hypothesis is based on the toxicity of beta A4 in cell cultures, and the findings that aggregation of beta A4 can be induced by metal-catalyzed oxidation and that free oxygen radicals might be involved in APP metabolism. Another neurological disorder, familial amyotrophic lateral sclerosis (FALS), supports our view that AD and FALS might be linked through a common mechanism. In FALS, SOD-Cu(I) complexes are affected by hydrogen peroxide and free radicals are produced. In AD, the reduction of Cu(II) to Cu(I) by APP involves an electron-transfer reaction and could also lead to a production of hydroxyl radicals. Thus, copper-mediated toxicity of APP-Cu(II)/(I) complexes may contribute to neurodegeneration in AD.
Collapse
Affiliation(s)
- G Multhaup
- ZMBH Center for Molecular Biology, University of Heidelberg, Germany
| |
Collapse
|
47
|
Beher D, Hesse L, Masters CL, Multhaup G. Regulation of amyloid protein precursor (APP) binding to collagen and mapping of the binding sites on APP and collagen type I. J Biol Chem 1996; 271:1613-20. [PMID: 8576160 DOI: 10.1074/jbc.271.3.1613] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The specific binding of the amyloid precursor protein (APP) to extracellular matrix molecules suggests that APP regulates cell interactions and has a function as a cell adhesion molecule and/or substrate adhesion molecule. On the molecular level APP has binding sites for collagen, laminin, and glycosaminoglycans which is a characteristic feature of cell adhesion molecules. We have examined the interactions between the APP and collagen types I and IV and identified the corresponding binding sites on APP and collagen type I. We show that APP bound most efficiently to collagen type I in a concentration-dependent and specific manner in the native and heat-denatured states, suggesting an involvement of a contiguous binding site on collagen. This binding site was identified on the cyanogen bromide fragment alpha 1(I)CB6 of collagen type I, which also binds heparin. APP did not bind to collagen type I-heparin complexes, which suggests that there are overlapping binding sites for heparin and APP on collagen. We localized the site of APP that mediates collagen binding within residues 448-465 of APP695, which are encoded by the ubiquitously expressed APP exon 12, whereas the high affinity heparin binding site of APP is located in exon 9. Since a peptide encompassing this region binds to collagen type I and inhibits APP-collagen type I binding in nanomolar concentrations, this region may comprise the major part of the collagen type I binding site of APP. Moreover, our data also indicate that the collagen binding site is involved in APP-APP interaction that can be modulated by Zn(II) and heparin. Taken together, the data suggest that the regulation of APP binding to collagen type I by heparin occurs through the competitive binding of heparin and APP to collagen.
Collapse
Affiliation(s)
- D Beher
- Center for Molecular Biology Heidelberg (ZMBH), University of Heidelberg, Federal Republic of Germany
| | | | | | | |
Collapse
|
48
|
Beyreuther K, Multhaup G, Mönning U, Sandbrink R, Beher D, Hesse L, Small DH, Masters CL. Regulation of APP expression, biogenesis and metabolism by extracellular matrix and cytokines. Ann N Y Acad Sci 1996; 777:74-6. [PMID: 8624129 DOI: 10.1111/j.1749-6632.1996.tb34403.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have identified and characterized the ligand binding properties of the Alzheimer's disease (AD) beta A4 amyloid protein precursor (APP), mapped the APP ligand binding sites and analyzed the regulation of APP expression, biogenesis and metabolism by components of the extracellular matrix (ECM) and cytokines.
Collapse
Affiliation(s)
- K Beyreuther
- ZMBH, Center for Molecular Biology, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Akaaboune M, Verdière-Sahuqué M, Lachkar S, Festoff BW, Hantaï D. Serine proteinase inhibitors in human skeletal muscle: expression of beta-amyloid protein precursor and alpha 1-antichymotrypsin in vivo and during myogenesis in vitro. J Cell Physiol 1995; 165:503-11. [PMID: 7593229 DOI: 10.1002/jcp.1041650308] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The balance of serine proteases and inhibitors in nerve and muscle is altered during programmed- and injury-induced remodeling. A serpin, alpha 1-antichymotrypsin (alpha 1-ACT), and Kunitz-inhibitor containing forms of the beta-amyloid precursor protein (beta APP) may be important components of this balance. In the present study, we analyzed their expression in primary cultures of human myogenic (satellite) cells that mimic myogenic differentiation using Western blotting and immunocytochemistry. In vitro results were compared to in vivo results from normal adult human skeletal muscle biopsies. Using an anti-alpha 1-ACT polyclonal antibody, we detected a 62 kDa immunoreactive band both in cultured human myogenic cells (mononucleated myoblasts as well as multi-nucleated myotubes) and in extracts of human muscle biopsies. With a polyclonal anti-beta APP antibody we found two bands (105 and 120 kDa) in myoblasts and myotubes in culture. However, the same antibody recognized only a single band at 92 kDa in biopsies. By immunocytochemistry, both alpha 1-ACT and beta APP were indistinctly present on localized to the surface of myoblasts in culture. In contrast, these inhibitors were dense on myotube surfaces, where they often formed distinct aggregates and frequently co-localized. In permeabilized muscle cells, alpha 1-ACT and beta APP appeared to be localized to the perikarya of both myoblasts and myotubes. Confirming previous results, both alpha 1-ACT and beta APP were present at the neuromuscular junction in human muscle sections. These developmental changes found during in vitro myogenesis for alpha 1-ACT and beta APP, both serine protease inhibitors, reinforce the hypothesis that regulation of the serine proteases and serine protease inhibitors plays an important role in neuromuscular differentiation.
Collapse
|
50
|
Abstract
The transition from short- to long-term memory requires lasting modulations of synaptic connectivity. In a variety of species and learning tasks, enhanced synthesis of glycoprotein cell-adhesion molecules (CAMs), such as neural CAM (NCAM) and Ll, 5-8 h post-training is a necessary step in this process. If the training event is weak, this phase of glycoprotein synthesis does not occur and memory is not retained. Antibodies or fragments that bind to the extracellular domains of NCAM or Ll at this time produce amnesia for the task. Centrally administered corticosterone enhances retention of weak learning, and steroid-receptor antagonists are amnestic. The effects of corticosterone are mediated through synthesis of 'second-wave' glycoproteins. As 'nootropic' drugs such as piracetam only enhance long-term retention and are ineffective in adrenalectomized animals, the interaction between glucocorticoids and glycoproteins might provide a site for pharmacological intervention in alleviating the losses of memory that occur in neurodegenerative disorders.
Collapse
Affiliation(s)
- S P Rose
- Brain and Behaviour Research Group, Open University, Milton Keynes, UK
| |
Collapse
|