1
|
Petralla S, Panayotova M, Franchina E, Fricker G, Puris E. Low-Density Lipoprotein Receptor-Related Protein 1 as a Potential Therapeutic Target in Alzheimer's Disease. Pharmaceutics 2024; 16:948. [PMID: 39065645 PMCID: PMC11279518 DOI: 10.3390/pharmaceutics16070948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease impacting the lives of millions of people worldwide. The formation of amyloid β (Aβ) plagues in the brain is the main pathological hallmark of AD. The Aβ deposits are formed due to the imbalance between the production and Aβ clearance in the brain and across the blood-brain barrier (BBB). In this respect, low-density lipoprotein receptor-related protein 1 (LRP1) plays a significant role by mediating both brain Aβ production and clearance. Due to its important role in AD pathogenesis, LRP1 is considered an attractive drug target for AD therapies. In the present review, we summarize the current knowledge about the role of LRP1 in AD pathogenesis as well as recent findings on changes in LRP1 expression and function in AD. Finally, we discuss the advances in utilizing LRP1 as a drug target for AD treatments as well as future perspectives on LRP1 research.
Collapse
Affiliation(s)
| | | | | | | | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany; (S.P.); (M.P.); (E.F.); (G.F.)
| |
Collapse
|
2
|
The transmembrane domain of the amyloid precursor protein is required for anti-amyloidogenic processing by α-secretase ADAM10. J Biol Chem 2022; 298:101911. [PMID: 35398353 PMCID: PMC9127328 DOI: 10.1016/j.jbc.2022.101911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
Neurotoxic amyloid β-peptides (Aβ) are thought to be a causative agent of Alzheimer's disease in humans. The production of Aβ from amyloid precursor protein (APP) could be diminished by enhancing α-processing; however, the physical interactions between APP and α-secretases are not well understood. In this study, we employed super-resolution light microscopy to examine in cell-free plasma membranes the abundance and association of APP and α-secretases ADAM10 and ADAM17. We found that both secretase molecules localize similarly closely to APP (within ≤ 50 nm). However, when cross-linking APP with antibodies directed against the GFP-tag of APP, in confocal microscopy we observed that only ADAM10 co-aggregated with APP. Furthermore, we mapped the involved protein domain by using APP variants with an exchanged transmembrane segment or lacking cytoplasmic/extracellular domains. We identified that APP's transmembrane domain is required for association with α-secretases and, as analysed by Western Blot, for α-processing. We propose that the APP transmembrane domain interacts either directly or indirectly with ADAM10, but not with ADAM17, explaining the dominant role of ADAM10 in α-processing of APP. Further understanding of this interaction may facilitate the development of a therapeutic strategy based on promoting APP cleavage by α-secretases.
Collapse
|
3
|
Mani S, Swargiary G, Singh M, Agarwal S, Dey A, Ojha S, Jha NK. Mitochondrial defects: An emerging theranostic avenue towards Alzheimer's associated dysregulations. Life Sci 2021; 285:119985. [PMID: 34592237 DOI: 10.1016/j.lfs.2021.119985] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/10/2021] [Accepted: 09/18/2021] [Indexed: 01/02/2023]
Abstract
Mitochondria play a crucial role in expediting the energy homeostasis under varying environmental conditions. As mitochondria are controllers of both energy production and apoptotic pathways, they are also distinctively involved in controlling the neuronal cell survival and/or death. Numerous factors are responsible for mitochondria to get degraded with aging and huge functional failures in mitochondria are also found to be associated with the commencement of numerous neurodegenerative conditions, including Alzheimer's disease (AD). A large number of existing literatures promote the pivotal role of mitochondrial damage and oxidative impairment in the pathogenesis of AD. Numerous mitochondria associated processes such as mitochondrial biogenesis, fission, fusion, mitophagy, transportation and bioenergetics are crucial for proper functioning of mitochondria but are reported to be defective in AD patients. Though, the knowledge on the precise and in-depth mechanisms of these actions is still in infancy. Based upon the outcome of various significant studies, mitochondria are also being considered as therapeutic targets for AD. Here, we review the current status of mitochondrial defects in AD and also summarize the possible role of these defects in the pathogenesis of AD. The various approaches for developing the mitochondria-targeted therapies are also discussed here in detail. Consequently, it is suggested that improving mitochondrial activity via pharmacological and/or non-pharmacological interventions could postpone the onset and slow the development of AD. Further research and consequences of ongoing clinical trials should extend our understanding and help to validate conclusions regarding the causation of AD.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201307, India.
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201307, India
| | - Manisha Singh
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, UP 201307, India
| | | | - Abhijit Dey
- Department of Life Sciences, Presidency University, College Street, Kolkata 700073, India
| | - Shreesh Ojha
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India
| |
Collapse
|
4
|
Oxford AE, Stewart ES, Rohn TT. Clinical Trials in Alzheimer's Disease: A Hurdle in the Path of Remedy. Int J Alzheimers Dis 2020; 2020:5380346. [PMID: 32308993 PMCID: PMC7152954 DOI: 10.1155/2020/5380346] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/18/2020] [Indexed: 11/17/2022] Open
Abstract
Human clinical trials seek to ameliorate the disease states and symptomatic progression of illnesses that, as of yet, are largely untreatable according to clinical standards. Ideally, clinical trials test "disease-modifying drugs," i.e., therapeutic agents that specifically modify pathological features or molecular bases of the disease and would presumably have a large impact on disease progression. In the case of Alzheimer's disease (AD), however, this approach appears to have stalled progress in the successful development of clinically useful therapies. For the last 25 years, clinical trials involving AD have centered on beta-amyloid (Aβ) and the Aβ hypothesis of AD progression and pathology. According to this hypothesis, the progression of AD begins following an accumulation of Aβ peptide, leading to eventual synapse loss and neuronal cell death: the true overriding pathological feature of AD. Clinical trials arising from the Aβ hypothesis target causal steps in the pathway in order to reduce the formation of Aβ or enhance clearance, and though agents have been successful in this aim, they remain unsuccessful in rescuing cognitive function or slowing cognitive decline. As such, further use of resources in the development of treatment options for AD that target Aβ, its precursors, or its products should be reevaluated. The purpose of this review was to give an overview of how human clinical trials are conducted in the USA and to assess the results of recent failed trials involving AD, the majority of which were based on the Aβ hypothesis. Based on these current findings, it is suggested that lowering Aβ is an unproven strategy, and it may be time to refocus on other targets for the treatment of this disease including pathological forms of tau.
Collapse
Affiliation(s)
- Alexandra E. Oxford
- Department of Biological Sciences, Boise State University, Science Building, Room 228, Boise, Idaho 83725, USA
| | - Erica S. Stewart
- Department of Biological Sciences, Boise State University, Science Building, Room 228, Boise, Idaho 83725, USA
| | - Troy T. Rohn
- Department of Biological Sciences, Boise State University, Science Building, Room 228, Boise, Idaho 83725, USA
| |
Collapse
|
5
|
Tang TC, Kienlen-Campard P, Hu Y, Perrin F, Opsomer R, Octave JN, Constantinescu SN, Smith SO. Influence of the familial Alzheimer's disease-associated T43I mutation on the transmembrane structure and γ-secretase processing of the C99 peptide. J Biol Chem 2019; 294:5854-5866. [PMID: 30755484 PMCID: PMC6463720 DOI: 10.1074/jbc.ra118.006061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/09/2019] [Indexed: 11/06/2022] Open
Abstract
Extracellular deposition of β-amyloid (Aβ) peptides in the brain is a hallmark of Alzheimer's disease (AD). Upon β-secretase-mediated cleavage of the β C-terminal fragment (β-CTF) from the Aβ precursor protein, the γ-secretase complex produces the Aβ peptides associated with AD. The familial T43I mutation within the transmembrane domain of the β-CTF (also referred to as C99) increases the ratio between the Aβ42 and Aβ40 peptides largely due to a decrease in Aβ40 formation. Aβ42 is the principal component of amyloid deposits within the brain parenchyma, and an increase in the Aβ42/Aβ40 ratio is correlated with early-onset AD. Using NMR and FTIR spectroscopy, here we addressed how the T43I substitution influences the structure of C55, the minimal sequence containing the entire extracellular and transmembrane (TM) domains of C99 needed for γ-secretase processing. 13C NMR chemical shifts indicated that the T43I substitution increases helical structure within the TM domain of C55. These structural changes were associated with a shift of the C55 dimer to the monomer and an increase in the tilt of the TM helix relative to the membrane normal in the T43I mutant compared with that of WT C55. The A21G (Flemish) mutation was previously found to increase secreted Aβ40 levels; here, we combined this mutation in the extracellular domain of C99 with T43I and observed that the T43I/A21G double mutant decreases Aβ40 formation. We discuss how the observed structural changes in the T43I mutant may decrease Aβ40 formation and increase the Aβ42/Aβ40 ratio.
Collapse
Affiliation(s)
- Tzu-Chun Tang
- From the Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215
| | | | - Yi Hu
- From the Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215
| | - Florian Perrin
- Ludwig Institute for Cancer Research and de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Rémi Opsomer
- the Institute of Neuroscience, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Jean-Noël Octave
- the Institute of Neuroscience, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research and de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Steven O Smith
- From the Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215.
| |
Collapse
|
6
|
Qian M, Shen X, Wang H. The Distinct Role of ADAM17 in APP Proteolysis and Microglial Activation Related to Alzheimer's Disease. Cell Mol Neurobiol 2016; 36:471-82. [PMID: 26119306 PMCID: PMC11482503 DOI: 10.1007/s10571-015-0232-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/23/2015] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with the symptom of cognitive impairment. The deposition of amyloid β (Aβ) peptide is believed to be the primary cause to neuronal dystrophy and eventually dementia. Aβ is the proteolytic product from its precursor amyloid precursor protein (APP) by β- and γ- secretase. An optional cleavage by α-secretase happens inside the Aβ domain. ADAM17 is supposed to be the regulated α-secretase of APP. Enhanced activity of ADAM17 leads to the increasing secretion of neuroprotective soluble APP α fragment and reduction of Aβ generation, which may be benefit to the disease. ADAM17 is then considered the potential therapeutic target for AD. Microglia activation and neuroinflammation is another important event in AD pathogenesis. Interestingly, ADAM17 also participates in the cleavage of many other membrane-bound proteins, especially some inflammatory factors related to microglia activation. The facilitating role of ADAM17 in inflammation and further neuronal damage has also been illustrated. In results, the activation of ADAM17 as the solution to AD may be a tricky task. The comprehensive consideration and evaluation has to be carried out carefully before the final treatment. In the present review, the distinct role of ADAM17 in AD-related APP shedding and neuroinflammatory microglial activation will be carefully discussed.
Collapse
Affiliation(s)
- Meng Qian
- Key Lab of Inflammation and Immunoregulation, School of Medicine, Hangzhou Normal University, Xuelin Street 16, Hangzhou, 310036, China
| | - Xiaoqiang Shen
- Key Lab of Inflammation and Immunoregulation, School of Medicine, Hangzhou Normal University, Xuelin Street 16, Hangzhou, 310036, China
| | - Huanhuan Wang
- Key Lab of Inflammation and Immunoregulation, School of Medicine, Hangzhou Normal University, Xuelin Street 16, Hangzhou, 310036, China.
| |
Collapse
|
7
|
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: 1.4] [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
|
8
|
Chen M. The Maze of APP Processing in Alzheimer's Disease: Where Did We Go Wrong in Reasoning? Front Cell Neurosci 2015; 9:186. [PMID: 26052267 PMCID: PMC4447002 DOI: 10.3389/fncel.2015.00186] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/28/2015] [Indexed: 02/04/2023] Open
Abstract
Why has Alzheimer’s disease (AD) remained a conundrum today? The main reason is the stagnation in understanding the origins of plaques and tangles. While they are widely thought to be the products of the “aberrant” pathways, we believe that plaques and tangles result from natural aging. From this new perspective, we have proposed that age-related inefficiency of α-secretase is the underpinning for Aβ overproduction. This view contrasts sharply with the current doctrine that Aβ overproduction is the product of the “overactivated” β- and γ-secretases. Following this doctrine, it has been claimed that the two secretases are “positively identified” and that their inhibitors have “successfully reduced Aβ levels.” But, why have these studies not led to the understanding of AD or successful clinical trials? And if so, where did they go off course in reasoning? These questions may touch the basics of biological science and must be answered. In this paper, I dissected several prevailing assumptions and some influential reports with an attempt to trace the origins of the conundrum. This work led me to an original model for Aβ overproduction and also to a serious question: given the universal knowledge that boosting α-secretase reduces Aβ, a straightforward highway for intervention, then why is there such an obsession on “inhibiting β- and γ-secretases,” a much more costly and twisting road even if possible? This issue requires the attention of policymakers and all researchers. I therefore call for a game change in AD study.
Collapse
Affiliation(s)
- Ming Chen
- Aging Research Laboratory, Research and Development Service, Bay Pines VA Healthcare System , Bay Pines, FL , USA ; Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, FL , USA
| |
Collapse
|
9
|
Affiliation(s)
- Brian H. Anderton
- Department of Neuroscience, Institute of Psychiatry, University of London, London, U.K
| |
Collapse
|
10
|
Rose C, Peoc'h K, Chasseigneaux S, Paquet C, Dumurgier J, Bourasset F, Calon F, Laplanche JL, Hugon J, Allinquant B. New highly sensitive rodent and human tests for soluble amyloid precursor protein alpha quantification: preclinical and clinical applications in Alzheimer's disease. BMC Neurosci 2012; 13:84. [PMID: 22824057 PMCID: PMC3418197 DOI: 10.1186/1471-2202-13-84] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/23/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amyloid precursor protein (APP), a key molecule in Alzheimer's disease (AD), is metabolized in two alternative cleavages, generating either the amyloidogenic peptides involved in AD pathology or the soluble form of APP (sAPPα). The level of amyloidogenic peptides in human cerebrospinal fluid (CSF) is considered to be a biomarker of AD, whereas the level of sAPPα in CSF as a biomarker has not been clearly established. sAPPα has neurotrophic and neuroprotective properties. Stimulating its formation and secretion is a promising therapeutic target in AD research. To this end, very sensitive tests for preclinical and clinical research are required. METHODS The tests are based on homogenous time-resolved fluorescence and require no washing steps. RESULTS We describe two new rapid and sensitive tests for quantifying mouse and human sAPPα. These 20 μl-volume tests quantify the levels of: i) endogenous mouse sAPPα in the conditioned medium of mouse neuron primary cultures, as well as in the CSF of wild-type mice, ii) human sAPPα in the CSF of AD mouse models, and iii) human sAPPα in the CSF of AD and non-AD patients. These tests require only 5 μl of conditioned medium from 5 × 10(4) mouse primary neurons, 1 μl of CSF from wild-type and transgenic mice, and 0.5 μl of human CSF. CONCLUSIONS The high sensitivity of the mouse sAPPα test will allow high-throughput investigations of molecules capable of increasing the secretion of endogenous sAPPα in primary neurons, as well as the in vivo validation of molecules of interest through the quantification of sAPPα in the CSF of treated wild-type mice. Active molecules could then be tested in the AD mouse models by quantifying human sAPPα in the CSF through the progression of the disease. Finally, the human sAPPα test could strengthen the biological diagnosis of AD in large clinical investigations. Taken together, these new tests have a wide field of applications in preclinical and clinical studies.
Collapse
Affiliation(s)
- Christiane Rose
- INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Schieb H, Weidlich S, Schlechtingen G, Linning P, Jennings G, Gruner M, Wiltfang J, Klafki HW, Knölker HJ. Structural design, solid-phase synthesis and activity of membrane-anchored β-secretase inhibitors on Aβ generation from wild-type and Swedish-mutant APP. Chemistry 2011; 16:14412-23. [PMID: 21132705 DOI: 10.1002/chem.201002878] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Covalent coupling of β-secretase inhibitors to a raftophilic lipid anchor via a suitable spacer by using solid-phase peptide synthesis leads to tripartite structures displaying substantially improved inhibition of cellular secretion of the β-amyloid peptide (Aβ). Herein, we describe a series of novel tripartite structures, their full characterization by NMR spectroscopy and mass spectrometry, and the analysis of their biological activity in cell-based assays. The tripartite structure concept is applicable to different pharmacophores, and the potency in terms of β-secretase inhibition can be optimized by adjusting the spacer length to achieve an optimal distance of the inhibitor from the lipid bilayer. A tripartite structure containing a transition-state mimic inhibitor was found to be less potent on Aβ generation from Swedish-mutant amyloid precursor protein (APP) than from the wild-type protein. Moreover, our observations suggest that specific variants of Aβ are generated from wild-type APP but not from Swedish-mutant APP and are resistant to β-secretase inhibition. Efficient inhibition of Aβ secretion by tripartite structures in the absence of appreciable neurotoxicity was confirmed in a primary neuronal cell culture, thus further supporting the concept.
Collapse
Affiliation(s)
- Heinke Schieb
- Department of Psychiatry and Psychotherapy, University of Duisburg-Essen, LVR-Klinikum, Essen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Gough M, Parr-Sturgess C, Parkin E. Zinc metalloproteinases and amyloid Beta-Peptide metabolism: the positive side of proteolysis in Alzheimer's disease. Biochem Res Int 2010; 2011:721463. [PMID: 21152187 PMCID: PMC2989646 DOI: 10.1155/2011/721463] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/07/2010] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease is a neurodegenerative condition characterized by an accumulation of toxic amyloid beta- (Aβ-)peptides in the brain causing progressive neuronal death. Aβ-peptides are produced by aspartyl proteinase-mediated cleavage of the larger amyloid precursor protein (APP). In contrast to this detrimental "amyloidogenic" form of proteolysis, a range of zinc metalloproteinases can process APP via an alternative "nonamyloidogenic" pathway in which the protein is cleaved within its Aβ region thereby precluding the formation of intact Aβ-peptides. In addition, other members of the zinc metalloproteinase family can degrade preformed Aβ-peptides. As such, the zinc metalloproteinases, collectively, are key to downregulating Aβ generation and enhancing its degradation. It is the role of zinc metalloproteinases in this "positive side of proteolysis in Alzheimer's disease" that is discussed in the current paper.
Collapse
Affiliation(s)
- Mallory Gough
- Division of Biomedical and Life Sciences, School of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Catherine Parr-Sturgess
- Division of Biomedical and Life Sciences, School of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Edward Parkin
- Division of Biomedical and Life Sciences, School of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
13
|
Kimura N, Inoue M, Okabayashi S, Ono F, Negishi T. Dynein dysfunction induces endocytic pathology accompanied by an increase in Rab GTPases: a potential mechanism underlying age-dependent endocytic dysfunction. J Biol Chem 2009; 284:31291-302. [PMID: 19758999 DOI: 10.1074/jbc.m109.012625] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Growing evidence suggests that endocytic dysfunction is intimately involved in early stage Alzheimer disease pathology, such as the accumulation of beta-amyloid precursor protein in enlarged early endosomes. However, it remains unclear how endocytic dysfunction is induced in an age-dependent manner. Cytoplasmic dynein, a microtubule-based motor protein, interacts with another microtubule-associated protein, dynactin. The resulting dynein-dynactin complex mediates minus end-directed vesicle transport, including endosome trafficking. We have previously shown that the interaction between dynein-dynactin complexes is clearly attenuated in aged monkey brains, suggesting that dynein-mediated transport dysfunction exists in aged brains. Our immunohistochemical analyses revealed that age-dependent endocytic pathology was accompanied by an increase in Rab GTPases in aged monkey brains. Here, we demonstrated that siRNA-induced dynein dysfunction reproduced the endocytic pathology accompanied by increased Rab GTPases seen in aged monkey brains and significantly disrupted exosome release. Moreover, it also resulted in endosomal beta-amyloid precursor protein accumulation characterized by increased beta-site cleavage. These findings suggest that dynein dysfunction may underlie age-dependent endocytic dysfunction via the up-regulation of Rab GTPases. In addition, this vicious circle may worsen endocytic dysfunction, ultimately leading to Alzheimer disease pathology.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1-1 Hachimandai, Tsukuba-shi, Ibaraki 305-0843, Japan.
| | | | | | | | | |
Collapse
|
14
|
Eggert S, Midthune B, Cottrell B, Koo EH. Induced dimerization of the amyloid precursor protein leads to decreased amyloid-beta protein production. J Biol Chem 2009; 284:28943-52. [PMID: 19596858 DOI: 10.1074/jbc.m109.038646] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The amyloid precursor protein (APP) plays a central role in Alzheimer disease (AD) pathogenesis because sequential cleavages by beta- and gamma-secretase lead to the generation of the amyloid-beta (Abeta) peptide, a key constituent in the amyloid plaques present in brains of AD individuals. In several studies APP has recently been shown to form homodimers, and this event appears to influence Abeta generation. However, these studies have relied on APP mutations within the Abeta sequence itself that may affect APP processing by interfering with secretase cleavages independent of dimerization. Therefore, the impact of APP dimerization on Abeta production remains unclear. To address this question, we compared the approach of constitutive cysteine-induced APP dimerization with a regulatable dimerization system that does not require the introduction of mutations within the Abeta sequence. To this end we generated an APP chimeric molecule by fusing a domain of the FK506-binding protein (FKBP) to the C terminus of APP. The addition of the synthetic membrane-permeant drug AP20187 induces rapid dimerization of the APP-FKBP chimera. Using this system we were able to induce up to 70% APP dimers. Our results showed that controlled homodimerization of APP-FKBP leads to a 50% reduction in total Abeta levels in transfected N2a cells. Similar results were obtained with the direct precursor of beta-secretase cleavage, C99/SPA4CT-FKBP. Furthermore, there was no modulation of different Abeta peptide species after APP dimerization in this system. Taken together, our results suggest that APP dimerization can directly affect gamma-secretase processing and that dimerization is not required for Abeta production.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
15
|
Rajendran L, Schneider A, Schlechtingen G, Weidlich S, Ries J, Braxmeier T, Schwille P, Schulz JB, Schroeder C, Simons M, Jennings G, Knölker HJ, Simons K. Efficient inhibition of the Alzheimer's disease beta-secretase by membrane targeting. Science 2008; 320:520-3. [PMID: 18436784 DOI: 10.1126/science.1156609] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
beta-Secretase plays a critical role in beta-amyloid formation and thus provides a therapeutic target for Alzheimer's disease. Inhibitor design has usually focused on active-site binding, neglecting the subcellular localization of active enzyme. We have addressed this issue by synthesizing a membrane-anchored version of a beta-secretase transition-state inhibitor by linking it to a sterol moiety. Thus, we targeted the inhibitor to active beta-secretase found in endosomes and also reduced the dimensionality of the inhibitor, increasing its local membrane concentration. This inhibitor reduced enzyme activity much more efficiently than did the free inhibitor in cultured cells and in vivo. In addition to effectively targeting beta-secretase, this strategy could also be used in designing potent drugs against other membrane protein targets.
Collapse
Affiliation(s)
- Lawrence Rajendran
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstr. 108, 01307 Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ikin AF, Causevic M, Pedrini S, Benson LS, Buxbaum JD, Suzuki T, Lovestone S, Higashiyama S, Mustelin T, Burgoyne RD, Gandy S. Evidence against roles for phorbol binding protein Munc13-1, ADAM adaptor Eve-1, or vesicle trafficking phosphoproteins Munc18 or NSF as phospho-state-sensitive modulators of phorbol/PKC-activated Alzheimer APP ectodomain shedding. Mol Neurodegener 2007; 2:23. [PMID: 18067682 PMCID: PMC2211485 DOI: 10.1186/1750-1326-2-23] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 12/09/2007] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Shedding of the Alzheimer amyloid precursor protein (APP) ectodomain can be accelerated by phorbol esters, compounds that act via protein kinase C (PKC) or through unconventional phorbol-binding proteins such as Munc13-1. We have previously demonstrated that application of phorbol esters or purified PKC potentiates budding of APP-bearing secretory vesicles at the trans-Golgi network (TGN) and toward the plasma membrane where APP becomes a substrate for enzymes responsible for shedding, known collectively as alpha-secretase(s). However, molecular identification of the presumptive "phospho-state-sensitive modulators of ectodomain shedding" (PMES) responsible for regulated shedding has been challenging. Here, we examined the effects on APP ectodomain shedding of four phorbol-sensitive proteins involved in regulation of vesicular membrane trafficking of APP: Munc13-1, Munc18, NSF, and Eve-1. RESULTS Overexpression of either phorbol-sensitive wildtype Munc13-1 or phorbol-insensitive Munc13-1 H567K resulted in increased basal APP ectodomain shedding. However, in contrast to the report of Rossner et al (2004), phorbol ester-dependent APP ectodomain shedding from cells overexpressing APP and Munc13-1 wildtype was indistinguishable from that observed following application of phorbol to cells overexpressing APP and Munc13-1 H567K mutant. This pattern of similar effects on basal and stimulated APP shedding was also observed for Munc18 and NSF. Eve-1, an ADAM adaptor protein reported to be essential for PKC-regulated shedding of pro-EGF, was found to play no obvious role in regulated shedding of sAPPalpha. CONCLUSION Our results indicate that, in the HEK293 system, Munc13-1, Munc18, NSF, and EVE-1 fail to meet essential criteria for identity as PMES for APP.
Collapse
Affiliation(s)
- Annat F Ikin
- Farber Institute for Neurosciences of Thomas Jefferson University, 900 Walnut Street, Philadelphia, 19107, PA, USA
| | - Mirsada Causevic
- Farber Institute for Neurosciences of Thomas Jefferson University, 900 Walnut Street, Philadelphia, 19107, PA, USA
| | - Steve Pedrini
- Farber Institute for Neurosciences of Thomas Jefferson University, 900 Walnut Street, Philadelphia, 19107, PA, USA
| | - Lyndsey S Benson
- Farber Institute for Neurosciences of Thomas Jefferson University, 900 Walnut Street, Philadelphia, 19107, PA, USA
| | - Joseph D Buxbaum
- Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, 10029, NY, USA
| | | | | | | | | | - Robert D Burgoyne
- Physiological Laboratory, University of Liverpool, Crown St, Liverpool, L69 3BX, UK
| | - Sam Gandy
- Farber Institute for Neurosciences of Thomas Jefferson University, 900 Walnut Street, Philadelphia, 19107, PA, USA
- Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, 10029, NY, USA
| |
Collapse
|
17
|
Wischik CM, Harrington CR, Mukaetova-Ladinska EB, Novak M, Edwards PC, McArthur FK. Molecular characterization and measurement of Alzheimer's disease pathology: implications for genetic and environmental aetiology. CIBA FOUNDATION SYMPOSIUM 2007; 169:268-93; discussion 293-302. [PMID: 1490426 DOI: 10.1002/9780470514306.ch16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The neuropathological changes seen in Alzheimer's disease represent an interaction between the ageing process in which normal intellectual function is retained, and changes which are specifically associated with severe cognitive deterioration. Molecular analysis of these changes has tended to emphasize the distinction between neurofibrillary pathology, which is intracellular and highly correlated with cognitive deterioration, and the changes associated with the deposition of extracellular amyloid, which appears to be widespread in normal ageing. Extracellular amyloid deposits consist of fibrils composed of a short 42 amino acid peptide (beta/A4) derived by abnormal proteolysis from a much larger precursor molecule (APP). The recent demonstration of a mutation associated with APP in rare cases with familial dementia, neurofibrillary pathology in the hippocampus and atypical cortical Lewy body pathology raises the possibility that abnormal processing of APP could be linked directly with neurofibrillary pathology. Neurofibrillary tangles and neuritic plaques are sites of dense accumulation of pathological paired helical filaments (PHFs) which are composed in part of an antigenically modified form of the microtubule-associated protein tau. The average brain tissue content of PHFs measured biochemically does not increase in the course of normal ageing but increases 10-fold relative to age-matched controls in patients with Alzheimer's disease. There is also a substantial (three-fold) disease-related decline in normal soluble tau protein relative to age-matched controls. This intracellular redistribution of a protein essential for microtubule stability in cortico-cortical association circuits may play an important part in the molecular pathogenesis of dementia in Alzheimer's disease. The role of abnormal proteolysis of APP in this process remains to be elucidated. Immunohistochemical studies on renal dialysis cases have failed to detect evidence of neurofibrillary pathology related to aluminium accumulation in brain tissue. Nevertheless it needs to be seen whether more sensitive biochemical assays of neurofibrillary pathology can demonstrate evidence of an association with aluminium.
Collapse
Affiliation(s)
- C M Wischik
- University of Cambridge Clinical School, Department of Psychiatry, UK
| | | | | | | | | | | |
Collapse
|
18
|
Lau TL, Ambroggio EE, Tew DJ, Cappai R, Masters CL, Fidelio GD, Barnham KJ, Separovic F. Amyloid-beta peptide disruption of lipid membranes and the effect of metal ions. J Mol Biol 2005; 356:759-70. [PMID: 16403524 DOI: 10.1016/j.jmb.2005.11.091] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 11/24/2005] [Accepted: 11/29/2005] [Indexed: 11/20/2022]
Abstract
Beta-amyloid peptide (Abeta), which is cleaved from the larger trans-membrane amyloid precursor protein, is found deposited in the brain of patients suffering from Alzheimer's disease and is linked with neurotoxicity. We report the results of studies of Abeta1-42 and the effect of metal ions (Cu2+ and Zn2+) on model membranes using 31P and 2H solid-state NMR, fluorescence and Langmuir Blodgett monolayer methods. Both the peptide and metal ions interact with the phospholipid headgroups and the effects on the lipid bilayer and the peptide structure were different for membrane incorporated or associated peptides. Copper ions alone destabilise the lipid bilayer and induced formation of smaller vesicles but when Abeta1-42 was associated with the bilayer membrane copper did not have this effect. Circular dichroism spectroscopy indicated that Abeta1-42 adopted more beta-sheet structure when incorporated in a lipid bilayer in comparison to the associated peptide, which was largely unstructured. Incorporated peptides appear to disrupt the membrane more severely than associated peptides, which may have implications for the role of Abeta in disease states.
Collapse
Affiliation(s)
- Tong-Lay Lau
- School of Chemistry, The University of Melbourne, Melbourne, Vic. 3010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Xu F, Davis J, Miao J, Previti ML, Romanov G, Ziegler K, Van Nostrand WE. Protease nexin-2/amyloid beta-protein precursor limits cerebral thrombosis. Proc Natl Acad Sci U S A 2005; 102:18135-40. [PMID: 16330760 PMCID: PMC1312400 DOI: 10.1073/pnas.0507798102] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The amyloid beta-protein precursor (AbetaPP) is best known as the parent molecule to the amyloid beta-peptide that accumulates in the brains of patients with Alzheimer's disease. Secreted isoforms of AbetaPP that contain the Kunitz proteinase inhibitor domain are analogous to the previously identified cell-secreted proteinase inhibitor known as protease nexin-2 (PN2). Although PN2/AbetaPP is enriched in brain and in circulating blood platelets, little is understood of its physiological function and potential role in disease processes outside of amyloid beta-peptide generation. We hypothesized that the potent inhibition of certain procoagulant proteinases by PN2/AbetaPP, coupled with its abundance in platelets and brain, indicate that it may function to regulate cerebral thrombosis. Here we show that specific and modest 2-fold overexpression of PN2/AbetaPP in circulating platelets of transgenic mice caused a marked inhibition of thrombosis in vivo. In contrast, deletion of PN2/AbetaPP in AbetaPP gene knockout mice resulted in a significant increase in thrombosis. Similarly, platelet PN2/AbetaPP transgenic mice developed larger hematomas in experimental intracerebral hemorrhage, whereas AbetaPP gene knockout mice exhibited reduced hemorrhage size. These findings indicate that PN2/AbetaPP plays a significant role in regulating cerebral thrombosis and that modest increases in this protein can profoundly enhance cerebral hemorrhage.
Collapse
Affiliation(s)
- Feng Xu
- Department of Medicine, Stony Brook University, NY 11794, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Wang J, Ho L, Qin W, Rocher AB, Seror I, Humala N, Maniar K, Dolios G, Wang R, Hof PR, Pasinetti GM. Caloric restriction attenuates beta-amyloid neuropathology in a mouse model of Alzheimer's disease. FASEB J 2005; 19:659-61. [PMID: 15650008 DOI: 10.1096/fj.04-3182fje] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
This study was designed to explore the possibility that caloric restriction (CR) may benefit Alzheimer's disease (AD) by preventing beta-amyloid (Abeta) neuropathology pivotal to the initiation and progression of the disease. We report that a CR dietary regimen prevents Abeta peptides generation and neuritic plaque deposition in the brain of a mouse model of AD neuropathology through mechanisms associated with promotion of anti-amyloidogenic alpha-secretase activity. Study findings support existing epidemiological evidence indicating that caloric intake may influence risk for AD and raises the possibility that CR may be used in preventative measures aimed at delaying the onset of AD amyloid neuropathology.
Collapse
Affiliation(s)
- Jun Wang
- Neuroinflammation Research Laboratories, Department of Psychiatry, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Eggert S, Paliga K, Soba P, Evin G, Masters CL, Weidemann A, Beyreuther K. The proteolytic processing of the amyloid precursor protein gene family members APLP-1 and APLP-2 involves alpha-, beta-, gamma-, and epsilon-like cleavages: modulation of APLP-1 processing by n-glycosylation. J Biol Chem 2004; 279:18146-56. [PMID: 14970212 DOI: 10.1074/jbc.m311601200] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Amyloid precursor protein (APP) processing is of major interest in Alzheimer's disease research, since sequential cleavages by beta- and gamma-secretase lead to the formation of the 4-kDa amyloid Abeta protein peptide that accumulates in Alzheimer's disease brain. The processing of APP involves proteolytic conversion by different secretases leading to alpha-, beta-, gamma-, delta-, and epsilon-cleavages. Since modulation of these cleavages represents a rational therapeutic approach to control amyloid formation, its interference with the processing of the members of the APP gene family is of considerable importance. By using C-terminally tagged constructs of APLP-1 and APLP-2 and the untagged proteins, we have characterized their proteolytic C-terminal fragments produced in stably transfected SH-SY5Y cells. Pharmacological manipulation with specific protease inhibitors revealed that both homologues are processed by alpha- and gamma-secretase-like cleavages, and that their intracellular domains can be released by cleavage at epsilon-sites. APLP-2 processing appears to be the most elaborate and to involve alternative cleavage sites. We show that APLP-1 is the only member of the APP gene family for which processing can be influenced by N-glycosylation. Additionally, we were able to detect p3-like fragments of APLP-1 and p3-like and Abeta-like fragments of APLP-2 in the media of stably transfected SH-SY5Y cells.
Collapse
Affiliation(s)
- Simone Eggert
- Zentrum für Molekulare Biologie Heidelberg, ZMBH, INF 282, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
22
|
Froestl B, Steiner B, Müller WE. Enhancement of proteolytic processing of the β-amyloid precursor protein by hyperforin. Biochem Pharmacol 2003; 66:2177-84. [PMID: 14609742 DOI: 10.1016/j.bcp.2003.08.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We studied the effect of hyperforin, a component of St. John's wort (Hypericum perforatum) extracts, on the processing of the amyloid precursor protein (APP) in rat pheochromocytoma PC12 cells, stably transfected with human wildtype APP. We observed transiently increased release of secretory APP fragments upon hyperforin treatment. Unique features, like a strong reduction of intracellular APP and the time course of soluble APP release, distinguished the effects of hyperforin from those of alkalizing agents and phorbol esters, well known activators of secretory processing of APP. Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), a protonophore, induced an almost identical decrease in intracellular pH in PC12 cells as does hyperforin. Despite this, FCCP induced a less pronounced release of soluble APP fragments and only slightly reduced intracellular APP levels. These results suggest that hyperforin is an activator of secretory processing of APP with a novel mechanism of action not solely dependent on its effects on intracellular pH.
Collapse
Affiliation(s)
- Bettina Froestl
- Department of Pharmacology, Biocenter, University of Frankfurt, Marie-Curie-Str. 9, D-60439 Frankfurt, Germany
| | | | | |
Collapse
|
23
|
Liu F, Su Y, Li B, Zhou Y, Ryder J, Gonzalez-DeWhitt P, May PC, Ni B. Regulation of amyloid precursor protein (APP) phosphorylation and processing by p35/Cdk5 and p25/Cdk5. FEBS Lett 2003; 547:193-6. [PMID: 12860412 DOI: 10.1016/s0014-5793(03)00714-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The phosphorylation status of amyloid precursor protein (APP) at Thr668 is suggested to play a critical role in the proteolytic cleavage of APP, which generates either soluble APP(beta) (sAPP(beta)) and beta-amyloid peptide (Abeta), the major component of senile plaques in patient brains inflicted with Alzheimer's disease (AD), or soluble APP(alpha) (sAPP(alpha)) and a peptide smaller than Abeta. One of the protein kinases known to phosphorylate APP(Thr668) is cyclin-dependent kinase 5 (Cdk5). Cdk5 is activated by the association with its regulatory partner p35 or its truncated form, p25, which is elevated in AD brains. The comparative effects of p35 and p25 on APP(Thr668) phosphorylation and APP processing, however, have not been reported. In this study, we investigated APP(Thr668) phosphorylation and APP processing mediated by p35/Cdk5 and p25/Cdk5 in the human neuroblastoma cell line SH-SY5Y. Transient overexpression of p35 and p25 elicited distinct patterns of APP(Thr668) phosphorylation, specifically, p35 increasing the phosphorylation of both mature and immature APP, whereas p25 primarily elevated the phosphorylation of immature APP. Despite these differential effects on APP phosphorylation, both p35 and p25 overexpression enhanced the secretion of Abeta, sAPP(beta), as well as sAPP(alpha). These results confirm the involvement of Cdk5 in APP processing, and suggest that p35- and p25-mediated Cdk5 activities lead to discrete APP phosphorylation.
Collapse
Affiliation(s)
- Feng Liu
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of the central nervous system (CNS) characterized by progressive loss of memory and other cognitive skills. Neurons in the limbic and association cortices become progressively dysfunctional affecting almost all cognitive functions and memory. The PSI-regulated epsilon-secretase cleavage of type I transmembrane receptors controls production of transcriptionally active intracellular fragments (ICFs) suggesting that this cleavage is a key factor in surface-to-nucleus signal transduction and gene expression. Signal-induced gene expression mediates neuronal responses to environmental changes and is a key event in neuronal survival and synaptic function. Familial Alzheimer's Disease (FAD) mutations may interfere with nuclear signaling and transcription by interfering with the PS1/epsilon-secretase cleavage and production of transcriptionally active ICFs. This raises the possibility that, similar to polyglutamine induced neurodegeneration like Huntington's chorea, transcriptional abnormalities are involved in the development of FAD.
Collapse
Affiliation(s)
- Nikolaos K Robakis
- Department of Psychiatry, Fishberg Reserch Center for Neurobiology, Mount Sinai School of Medicine, New York University, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
25
|
Davis J, Wagner MR, Zhang W, Xu F, Van Nostrand WE. Amyloid beta-protein stimulates the expression of urokinase-type plasminogen activator (uPA) and its receptor (uPAR) in human cerebrovascular smooth muscle cells. J Biol Chem 2003; 278:19054-61. [PMID: 12754271 DOI: 10.1074/jbc.m301398200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The accumulation of fibrillar amyloid-beta protein (A beta) in cerebral blood vessels, a condition known as cerebral amyloid angiopathy (CAA), is a key pathological feature of Alzheimer's disease and certain related disorders and is intimately associated with cerebrovascular cell death both in vivo and in vitro. Moreover, severe CAA leads to loss of vessel wall integrity and cerebral hemorrhage. Although the basis for these latter pathological consequences in CAA remains unresolved alterations in local proteolytic mechanisms may be involved. Here we show that pathogenic forms of A beta stimulate the expression of plasminogen activator activity in cultured human cerebrovascular smooth muscle (HCSM) cells, an in vitro model of CAA. RNase protection assays and plasminogen zymography showed that urokinase-type plasminogen activator (uPA) was responsible for this activity. There was preferential accumulation of uPA on the HCSM cell surface that was mediated through a concomitant increase in expression of the uPA receptor. In the presence of plasminogen there was robust degradation of A beta that was added to the HCSM cells resulting in restoration of cell viability. This suggests that increased expression of uPA may initially serve as a protective mechanism leading to localized degradation and clearance of the pathogenic stimulus A beta. On the other hand, chronic expression of uPA and plasminogen activation led to a profound loss of HCSM cell attachment. This suggests that a similar prolonged effect in vivo in the cerebral vessel wall may contribute to loss of integrity and cerebral hemorrhage in CAA.
Collapse
Affiliation(s)
- Judianne Davis
- Department of Medicine, Health Sciences Center, Stony Brook University, Stony Brook, New York 11794-8153, USA
| | | | | | | | | |
Collapse
|
26
|
Van Nostrand WE, Melchor JP, Keane DM, Saporito-Irwin SM, Romanov G, Davis J, Xu F. Localization of a fibrillar amyloid beta-protein binding domain on its precursor. J Biol Chem 2002; 277:36392-8. [PMID: 12107175 DOI: 10.1074/jbc.m204676200] [Citation(s) in RCA: 42] [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
Deposition of fibrillar amyloid-beta protein (Abeta) in senile plaques and in the walls of cerebral blood vessels is a key pathological feature of Alzheimer's disease and certain related disorders. Fibrillar Abeta deposition is intimately associated with neuronal and cerebrovascular cell death both in vivo and in vitro. Similarly, accumulation of the Abeta protein precursor (AbetaPP) is also observed at sites of fibrillar Abeta deposition. Recently, we reported that fibrillar Abeta, but not unassembled Abeta, promotes the specific binding of AbetaPP through its cysteine-rich, amino-terminal region (Melchor, J. P., and Van Nostrand, W. E. (2000) J. Biol. Chem. 275, 9782-9791). In the present study we sought to determine the precise site on AbetaPP that facilitates its binding to fibrillar Abeta. A series of synthesized overlapping peptides spanning the cysteine-rich, amino-terminal region of AbetaPP were used as competitors for AbetaPP binding to fibrillar Abeta. A peptide spanning residues 105-119 of AbetaPP competitively inhibited AbetaPP binding to fibrillar Abeta in a solid-phase binding assay and on the surface of cultured human cerebrovascular smooth muscle cells. Alanine-scanning mutagenesis of residues 105-117 within glutathione S-transferase (GST)-AbetaPP-(18-119) revealed that His(110), Val(112), and Ile(113) are key residues that facilitate AbetaPP binding to fibrillar Abeta. These specific residues belong to a common beta-strand within this region of AbetaPP. Wild-type GST-AbetaPP-(18-119) protected cultured human cerebrovascular smooth muscle cells from Abeta-induced toxicity whereas H110A mutant GST-AbetaPP-(18-119) did not. Wild-type GST-AbetaPP-(18-119) bound to different isoforms of fibrillar Abeta and fibrillar amylin peptides whereas H110A mutant and I113A mutant GST-AbetaPP-(18-119) were substantially less efficient binding to each fibrillar peptide. We conclude that His(110), Val(112), and Ile(113), residing in a common beta-strand region within AbetaPP-(18-119), comprise a domain that mediates the binding of AbetaPP to fibrillar peptides.
Collapse
Affiliation(s)
- William E Van Nostrand
- Department of Medicine, Health Sciences Center, Stony Brook University, Stony Brook, New York 11794-8153, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Rao RR, Kisaalita WS. Biochemical and electrophysiological differentiation profile of a human neuroblastoma (IMR-32) cell line. In Vitro Cell Dev Biol Anim 2002; 38:450-6. [PMID: 12605539 DOI: 10.1290/1071-2690(2002)038<0450:baedpo>2.0.co;2] [Citation(s) in RCA: 13] [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
A human neuroblastoma cell line (IMR-32), when differentiated, mimics large projections of the human cerebral cortex and under certain tissue culture conditions, forms intracellular fibrillary material, commonly observed in brains of patients affected with Alzheimer's disease. Our purpose is to use differentiated IMR-32 cells as an in vitro system for magnetic field exposure studies. We have previously studied in vitro differentiation of murine neuroblastoma (N1E-115) cells with respect to resting membrane potential development. The purpose of this study was to extend our investigation to IMR-32 cells. Electrophysiological (resting membrane potential, V(m)) and biochemical (neuron-specific enolase activity [NSE]) measurements were taken every 2 d for a period of 16 d. A voltage-sensitive oxonol dye together with flow cytometry was used to measure relative changes in V(m). To rule out any effect due to mechanical cell detachment, V(m) was indirectly measured by using a slow potentiometric dye (tetramethylrhodamine methyl ester) together with confocal digital imaging microscopy. Neuron-specific enolase activity was measured by following the production of phosphoenolpyruvate from 2-phospho-d-glycerate at 240 nm. Our results indicate that in IMR-32, in vitro differentiation as characterized by an increase in NSE activity is not accompanied by resting membrane potential development. This finding suggests that pathways for morphological-biochemical and electrophysiological differentiations in IMR-32 cells are independent of one another.
Collapse
Affiliation(s)
- Raj R Rao
- Cellular Bioengineering Laboratory, Biological and Agricultural Engineering Department, Driftmier Engineering Center, University of Georgia, Athens 30602, USA
| | | |
Collapse
|
28
|
Beher D, Wrigley JDJ, Owens AP, Shearman MS. Generation of C-terminally truncated amyloid-beta peptides is dependent on gamma-secretase activity. J Neurochem 2002; 82:563-75. [PMID: 12153480 DOI: 10.1046/j.1471-4159.2002.00985.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aberrant production of amyloid-beta peptides by processing of the beta-amyloid precursor protein leads to the formation of characteristic extracellular protein deposits which are thought to be the cause of Alzheimer's disease. Therefore, inhibiting the key enzymes responsible for amyloid-beta peptide generation, beta- and gamma-secretase may offer an opportunity to intervene with the progression of the disease. In human brain and cell culture systems a heterogeneous population of amyloid-beta peptides with various truncations is detected and at present, it is unclear how they are produced. We have used a combination of surface enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS) and a specific inhibitor of gamma-secretase to investigate whether the production of all amyloid-beta peptide species requires the action of gamma-secretase. Using this approach, we demonstrate that the production of all truncated amyloid-beta peptides except those released by the action of the nonamyloidogenic alpha-secretase enzyme or potentially beta-site betaAPP cleaving enzyme 2 depends on gamma-secretase activity. This indicates that none of these peptides are generated by a separate enzyme entity and a specific inhibitor of the gamma-secretase enzyme should havethe potential to block the generation of all amyloidogenicpeptides. Furthermore in the presence of gamma-secretase inhibitors, the observation of increased cleavage of the membrane-bound betaAPP C-terminal fragment C99 by alpha-secretase suggests that during its trafficking C99 encounters compartments in which alpha-secretase activity resides.
Collapse
Affiliation(s)
- Dirk Beher
- Department of Biochemistry & Molecular Biology, Merck Sharp & Dohme Research Laboratories, Harlow, Essex, UK.
| | | | | | | |
Collapse
|
29
|
Abstract
Biochemical and genetic evidence indicates the balance of biogenesis/clearance of Abeta amyloid peptides is altered in Alzheimer's disease. Abeta is derived, by two sequential cleavages, from the receptor-like amyloid precursor protein (APP). The proteases involved are beta-secretase, identified as the novel aspartyl protease BACE, and gamma-secretase, a multimeric complex containing the presenilins (PS). Gamma-secretase can release either Abeta40 or the more aggregating and cytotoxic Abeta42. Secreted Abeta peptides become either degraded by the metalloproteases insulin-degrading enzyme (IDE) and neprilysin or metabolized through receptor uptake mediated by apolipoprotein E. Therapeutic approaches based on secretase inhibition or amyloid clearance are currently under development.
Collapse
Affiliation(s)
- Geneviève Evin
- Department of Pathology, The University of Melbourne and Mental Health Research Institute, Parkville, Vic. 3010, Australia.
| | | |
Collapse
|
30
|
Sambamurti K, Hardy J, Refolo LM, Lahiri DK. Targeting APP metabolism for the treatment of Alzheimer's disease. Drug Dev Res 2002. [DOI: 10.1002/ddr.10077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
31
|
Sisodia SS, St George-Hyslop PH. gamma-Secretase, Notch, Abeta and Alzheimer's disease: where do the presenilins fit in? Nat Rev Neurosci 2002; 3:281-90. [PMID: 11967558 DOI: 10.1038/nrn785] [Citation(s) in RCA: 415] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sangram S Sisodia
- Center for Molecular Neurobiology, Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
32
|
van den Hurk WH, Bloemen M, Martens GJ. Expression of the gene encoding the beta-amyloid precursor protein APP in Xenopus laevis. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 97:13-20. [PMID: 11744158 DOI: 10.1016/s0169-328x(01)00279-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The beta-amyloid precursor protein APP is generally accepted to be directly or indirectly involved in the neurodegenerative disorder Alzheimer's disease and has been extensively studied in a number of mammalian systems. Its normal function remains, however, still elusive. We have used the clawed toad, Xenopus laevis, to study the first non-mammalian APP protein. Screening of a Xenopus laevis intermediate pituitary cDNA library led to the identification of two structurally different APP gene transcripts presumably resulting from duplicated genes. Sequence comparison between the Xenopus and human APP proteins revealed at the amino acid sequence level an identity of 92%. Both Xenopus genes were found to be expressed in all tissues examined, but their expression levels differed among tissues. In addition, as in mammals, alternative splicing was observed and the alternatively spliced APP(695) mRNA variant was expressed predominantly in the brain and the oocyte, while the longer isoforms (APP(751-770)) were predominant in the other tissues examined. Of special interest is the finding that, like human but unlike mouse or rat beta-amyloid (Abeta), the Xenopus peptide contains all amino acid residues implicated in amyloidogenesis. We conclude that Xenopus APP mRNA is ubiquitously expressed and alternatively spliced, and that the highly conserved Xenopus APP protein contains an Abeta peptide with amyloidogenic potency.
Collapse
Affiliation(s)
- W H van den Hurk
- Department of Molecular Animal Physiology, University of Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | | | | |
Collapse
|
33
|
Abstract
The amyloid precursor protein (APP) gene and its protein products have multiple functions in the central nervous system and fulfil criteria as neuractive peptides: presence, release and identity of action. There is increased understanding of the role of secretases (proteases) in the metabolism of APP and the production of its peptide fragments. The APP gene and its products have physiological roles in synaptic action, development of the brain, and in the response to stress and injury. These functions reveal the strategic importance of APP in the workings of the brain and point to its evolutionary significance.
Collapse
Affiliation(s)
- P K Panegyres
- Department of Neuropathology, Royal Perth Hospital, Western Australia.
| |
Collapse
|
34
|
Gitter BD, Boggs LN, May PC, Czilli DL, Carlson CD. Regulation of cytokine secretion and amyloid precursor protein processing by proinflammatory amyloid beta (A beta). Ann N Y Acad Sci 2001; 917:154-64. [PMID: 11268339 DOI: 10.1111/j.1749-6632.2000.tb05379.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neurodegenerative processes in Alzheimer's disease (AD) are thought to be driven, in part, by the deposition of amyloid beta (A beta), a 39-43-aminoacid peptide product resulting from an alternative cleavage of amyloid precursor protein (APP). In addition to its neurotoxic properties, A beta may influence neuropathology by stimulating glial cell cytokine and acute phase protein secretion in affected areas of the brain (e.g., cortex, hippocampus). Using an in vitro human astrocyte model (U-373 MG astrocytoma cells), the effects of A beta treatment on acute phase protein (APP and alpha-1-antichymotrypsin [alpha 1-ACT]) and interleukin-8 (IL-8) were examined. U-373 MG cells secreted increased levels of alpha 1-ACT and neurotrophic/neuroprotective alpha-cleaved APP (alpha APP) after exposure to interleukin-1 beta (IL-1 beta) for 24 hours. A beta treatment resulted in a similar, but modest increase in alpha 1-ACT secretion, a two- to threefold stimulation of IL-8 production, and, conversely, a profound reduction in the levels of secreted alpha APPs. A beta inhibited alpha APP secretion by U-373 MG cells in a concentration- and conformation-dependent manner. Moreover, the reduction in alpha APP secretion was accompanied by an increase in cell-associated APP. Another proinflammatory amyloidogenic peptide, human amylin, similarly affected APP processing in U-373 astrocytoma cells. These data suggest that A beta may contribute to Alzheimer's-associated neuropathology by lowering the production of neuroprotective/neurotrophic alpha APPs. Moreover, the concomitant increase in cell-associated APP may provide increased substrate for the generation of amyloidogenic peptides within astrocytes.
Collapse
Affiliation(s)
- B D Gitter
- Neuroscience Diseases Research Division, Lilly Research Laboratories, Eli Lilly and Co., Lilly Corporate Center, Indianapolis, Indiana 46285, USA.
| | | | | | | | | |
Collapse
|
35
|
Farzan M, Schnitzler CE, Vasilieva N, Leung D, Choe H. BACE2, a beta -secretase homolog, cleaves at the beta site and within the amyloid-beta region of the amyloid-beta precursor protein. Proc Natl Acad Sci U S A 2000; 97:9712-7. [PMID: 10931940 PMCID: PMC16930 DOI: 10.1073/pnas.160115697] [Citation(s) in RCA: 294] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Production of amyloid-beta protein (Abeta) is initiated by a beta-secretase that cleaves the Abeta precursor protein (APP) at the N terminus of Abeta (the beta site). A recently identified aspartyl protease, BACE, cleaves the beta site and at residue 11 within the Abeta region of APP. Here we show that BACE2, a BACE homolog, cleaves at the beta site and more efficiently at a different site within Abeta. The Flemish missense mutation of APP, implicated in a form of familial Alzheimer's disease, is adjacent to this latter site and markedly increases Abeta production by BACE2 but not by BACE. BACE and BACE2 respond identically to conservative beta-site mutations, and alteration of a common active site Arg inhibits beta-site cleavage but not cleavage within Abeta by both enzymes. These data suggest that BACE2 contributes to Abeta production in individuals bearing the Flemish mutation, and that selective inhibition of these highly similar proteases may be feasible and therapeutically advantageous.
Collapse
Affiliation(s)
- M Farzan
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
36
|
Salinero O, Moreno-Flores MT, Wandosell F. Increasing neurite outgrowth capacity of beta-amyloid precursor protein proteoglycan in Alzheimer's disease. J Neurosci Res 2000; 60:87-97. [PMID: 10723071 DOI: 10.1002/(sici)1097-4547(20000401)60:1<87::aid-jnr9>3.0.co;2-c] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Progressive cerebral deposition of beta-amyloid peptide either in blood vessels or around neurites is one of the most important features of Alzheimer's disease (AD). The beta-peptide, known as Abeta or A4, is produced by proteolytic cleavage of the amyloid precursor protein (APP). Two APP processing pathways have been proposed as physiological alternatives; only one of which leads to the production of Abeta or amyloidogenic peptides. However, we have little information regarding these processing pathways in the brain, or on whether posttranslational modifications such as glycosylation affect APP processing in vivo. Furthermore, the physiological function(s) of this protein in nervous tissue remains unclear, although modulatory roles in cell adhesion and neuritic extension have been suggested. It has been reported that APP may be glycosylated as a proteoglycan. We purified this APP population from human brain, and our data indicate that PG-APP supports neurite extension of hippocampal neurons. Neurons grown on this substratum showed an increased capacity to elongate neurites and increased neuritic "branching" compared to culture on laminin. These effects were enhanced with PG-APP samples obtained from AD brains. Our results suggest that this APP population may act as a neurite outgrowth and branching promoter and may thus play a role in some pathological conditions. These findings may have significant implications in understanding normal brain development and pathological situations (such as AD).
Collapse
Affiliation(s)
- O Salinero
- Centro de Biología Molecular "Severo Ochoa," CSIC-Universidad Autonoma de Madrid, Cantoblanco-Madrid, Spain
| | | | | |
Collapse
|
37
|
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: 29] [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
|
38
|
Yuan H, Zhai P, Anderson LM, Pan J, Thimmapaya B, Koo EH, Marquez-Sterling NR. Recombinant adenovirus is an appropriate vector for endocytotic protein trafficking studies in cultured neurons. J Neurosci Methods 1999; 88:45-54. [PMID: 10379578 DOI: 10.1016/s0165-0270(99)00011-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Endocytosis of full-length beta-amyloid precursor protein (APP) from the plasma membrane contributes to beta-amyloid peptide (Abeta) secretion, and, hence, potentially contributes to the molecular pathogenesis of Alzheimer's disease. We recently have demonstrated that central neuronal APP is endocytosed in a common vesicular compartment with recycling synaptic vesicle integral membrane proteins, but is then sorted away from synaptic vesicles for retrograde transport to the neuronal soma. For this report, we explore whether recombinant adenovirus can be used to modulate APP expression in cultured central neurons to study APP processing by the endocytotic pathway in these cells. Using a replication-deficient recombinant adenovirus that expresses a lacZ reporter (Ad5/CMV-lacZ), we demonstrate high efficiency of transfection (30-35%) at low viral titer (10-20 MOI), with no significant neuronal toxicity or cytoarchitectural change. In addition, we demonstrate that infection with the control virus does not result in re-direction of endogenous neuronal APP from usual endocytotic pathways. We have prepared, using the same genomic background as the control virus, an adenoviral vector that expresses the neuronal isoform of human APP (Ad5/CMV-APP). Infection with Ad5/CMV-APP at 10-20 MOI results in significantly increased immunoreactivity for endocytosed APP with preservation of usual endocytotic trafficking. These results demonstrate that recombinant adenovirus at low titer is an appropriate and effective vector for protein trafficking/processing studies in cultured central neurons.
Collapse
Affiliation(s)
- H Yuan
- Department of Pathology, Northwestern University Medical School, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Wagey RT, Krieger C. Abnormalities of protein kinases in neurodegenerative diseases. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 1999; 51:133-83. [PMID: 9949861 DOI: 10.1007/978-3-0348-8845-5_4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In neurodegenerative diseases such as ALS and AD there is evidence for abnormal regulation of protein kinases. In these diseases, altered activities and protein levels of several specific kinases suggest that abnormal phosphorylation is present and this aberrant phosphorylation may be involved in the pathogenesis of these diseases. The observation that regulation of the NMDA receptor ion channel is altered in tissue from ALS patients may arise from the abnormal phosphorylation state of the protein kinase regulating NMDA receptor function. Whether the abnormalities of these protein kinases is a primary event leading to altered receptor regulation or vice versa is still poorly understood. The seemingly multiple pathogenic mechanisms of ALS and AD create complexity in assessing a primary cause that may lead to cell death. The mechanisms causing cell death (apoptosis or necrosis) may be overlapping with integrated events among the components interacting and contributing to a final pathway for neuron death. Thus, evidence of impairment in protein kinase signalling in these diseases may be a primary cause, a secondary event, or a compensatory mechanism. To further study this issue, different model systems could be beneficial to obtain a better understanding of these diseases.
Collapse
Affiliation(s)
- R T Wagey
- Dept. of Medicine, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
40
|
Abstract
Multiple lines of evidence suggest that increased production and/or deposition of the beta-amyloid peptide, derived from the amyloid precursor protein, contributes to Alzheimer's disease. A growing list of neurotransmitters, growth factors, cytokines, and hormones have been shown to regulate amyloid precursor protein processing. Although traditionally thought to be mediated by activation of protein kinase C, recent data have implicated other signaling mechanisms in the regulation of this process. Moreover, novel mechanisms of regulation involving cholesterol-, apolipoprotein E-, and stress-activated pathways have been identified. As the phenotypic changes associated with Alzheimer's disease encompass many of these signaling systems, it is relevant to determine how altered cell signaling may be contributing to increasing brain amyloid burden. We review the myriad ways in which first messengers regulate amyloid precursor protein catabolism as well as the signal transduction cascades that give rise to these effects.
Collapse
Affiliation(s)
- J Mills
- Kinsmen Laboratory of Neurological Research, Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
41
|
Borchelt DR, Wong PC, Sisodia SS, Price DL. Transgenic mouse models of Alzheimer's disease and amyotrophic lateral sclerosis. Brain Pathol 1998; 8:735-57. [PMID: 9804381 PMCID: PMC8098285 DOI: 10.1111/j.1750-3639.1998.tb00198.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the past several years, there has been enormous progress in generating transgenic mice that model aspects of human neurodegenerative diseases. These studies build upon the efforts of molecular geneticists who have identified a number of genes that, when mutated, cause familial forms of these diseases. In this review, we focus on the mutations that cause familial forms of Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS), and transgenic mouse models that develop clinical and pathological abnormalities resembling those occurring in the human diseases.
Collapse
Affiliation(s)
- D R Borchelt
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
42
|
Salinero O, Garrido JJ, Wandosell F. Amyloid precursor protein proteoglycan is increased after brain damage. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1406:237-50. [PMID: 9630651 DOI: 10.1016/s0925-4439(98)00009-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The beta-amyloid peptide (Abeta or A4) is produced by proteolytic cleavage from amyloid precursor protein (APP). The progressive cerebral deposition of this peptide is one of the most important features of Alzheimer's disease. From the study of normal and transfected cells, two APP processing pathways have been proposed as physiological alternatives. One of these can produce Abeta or amyloidogenic peptides, whereas the second does not. However, it is not completely clear how APPs are post-translationally modified, proteolytically processed and metabolized in the brain. We report here that APPs also exist as proteoglycan, chondroitin-sulfate (ChS). We have identified in normal rat brain a complex pool of 8 to 130 kDa ChS-core proteins. The main portion of these proteoglycan (PGs) APPs contains complete amyloidogenic sequence, suggesting a novel proteolytic processing of APP from the amino-terminal to the transmembrane region. This population appears augmented after brain damage. These findings may have significant implications in understanding the initial deposition and kinetics of amyloid aggregation in a pathological situation like Alzheimer's disease.
Collapse
Affiliation(s)
- O Salinero
- Centro de Biología Molecular 'Severo Ochoa', CSIC-Universidad Autonoma de Madrid, Cantoblanco-Madrid 28049, Spain
| | | | | |
Collapse
|
43
|
Ikezu T, Trapp BD, Song KS, Schlegel A, Lisanti MP, Okamoto T. Caveolae, plasma membrane microdomains for alpha-secretase-mediated processing of the amyloid precursor protein. J Biol Chem 1998; 273:10485-95. [PMID: 9553108 DOI: 10.1074/jbc.273.17.10485] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Caveolae are plasma membrane invaginations where key signaling elements are concentrated. In this report, both biochemical and histochemical analyses demonstrate that the amyloid precursor protein (APP), a source of Abeta amyloid peptide, is enriched within caveolae. Caveolin-1, a principal component of caveolae, is physically associated with APP, and the cytoplasmic domain of APP directly participates in this binding. The characteristic C-terminal fragment that results from APP processing by alpha-secretase, an as yet unidentified enzyme that cleaves APP within the Abeta amyloid sequence, was also localized within these caveolae-enriched fractions. Further analysis by cell surface biotinylation revealed that this cleavage event occurs at the cell surface. Importantly, alpha-secretase processing was significantly promoted by recombinant overexpression of caveolin in intact cells, resulting in increased secretion of the soluble extracellular domain of APP. Conversely, caveolin depletion using antisense oligonucletotides prevented this cleavage event. Our current results indicate that caveolae and caveolins may play a pivotal role in the alpha-secretase-mediated proteolysis of APP in vivo.
Collapse
Affiliation(s)
- T Ikezu
- Department of Neurosciences, The Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
The most common cause of dementia occurring in mid- to late-life is Alzheimer's disease (AD). Some cases of AD, particularly those of early onset, are familial and inherited as autosomal dominant disorders linked to the presence of mutant genes that encode the amyloid precursor protein (APP) or the presenilins (PS1 or PS2). These mutant gene products cause dysfunction/death of vulnerable populations of nerve cells important in memory, higher cognitive processes, and behavior. AD affects 7-10% of individuals > 65 years of age and perhaps 40% of individuals > 80 years of age. For the late-onset cases, the principal risk factors are age and apolipoprotein (apoE) allele type, with apoE4 allele being a susceptibility factor. In this review, we briefly discuss the clinical syndrome of AD and the neurobiology/neuropathology of the disease and then focus attention on mutant genes linked to autosomal dominant familial AD (FAD), the biology of the proteins encoded by these genes, and the recent exciting progress in investigations of genetically engineered animal models that express these mutant genes and develop some features of AD.
Collapse
Affiliation(s)
- D L Price
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2196, USA. ,
| | | |
Collapse
|
45
|
Abstract
The genetic associations with the pathological features of AD are diverse: A rapidly growing number of mutations in presenilin 1 and 2 on chromosomes 14 and 1, respectively, are found in many early-onset FAD patients (Lendon et al., 1997). In addition, beta PP mutations are found in a small percentage of early-onset FAD kindreds. The apoE4 allele on chromosome 19 is associated with the presence of the most common form of AD, sporadic AD (Wisniewski & Frangione, 1992; Namba et al., 1991). However, it is clear that other proteins are also involved in the pathogenesis of AD, since some early-onset FAD kindreds do not have linkage to PS1, PS2, apoE, or beta PP, while at least 50% of late-onset AD is unrelated to apoE. Other proteins which have been implicated in the formation of senile plaques, but so far are not known to have any genetic linkage to AD, include proteoglycans (Snow et al., 1987), apoA1 (Wisniewski et al., 1995a), alpha 1-antichymotrypsin (Abraham et al., 1988), HB-GAM (Wisniewski et al., 1996a), complement components (McGeer & Rogers, 1992), acetylcholinesterase (Friede, 1965), and NAC (Ueda et al., 1993). Which of these proteins will be the most important for the etiology of the most common form of AD, late-onset sporadic AD, remains an open question. Three of the genes which are now known to be linked to AD, including PS1, beta PP, and apoE, have been established immunohistochemically and biochemically to be components of senile plaques (see Fig. 1). This raises at least two possibilities: either each of these proteins is part of one pathway with A beta-related amyloid formation as a final causative pathogenic event or amyloid deposition in AD is a reactive process related to dysfunction of a number of different CNS proteins. Whether or not amyloid formation is directly causative in the pathogenesis of AD, current data suggest that new therapeutic approaches which may inhibit the aggregation and/or the conformational change of sA beta to A beta fibrils (Soto et al., 1996) have the greatest likelihood to make a significant impact on controlling amyloid accumulation in AD.
Collapse
Affiliation(s)
- T Wisniewski
- Department of Neurology, New York University Medical Center, New York 10016, USA.
| | | | | |
Collapse
|
46
|
Wong PC, Borchelt DR, Lee MK, Pardo CA, Thinakaran G, Martin LJ, Sisodia SS, Price DL. Familial Amyotrophic Lateral Sclerosis and Alzheimer’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1998. [DOI: 10.1007/978-1-4615-4869-0_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
|
47
|
Haas C, Aldudo J, Cazorla P, Bullido MJ, de Miguel C, Vázquez J, Valdivieso F. Proteolysis of Alzheimer's disease beta-amyloid precursor protein by factor Xa. BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1343:85-94. [PMID: 9428662 DOI: 10.1016/s0167-4838(97)00094-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Amyloid beta-protein is a 4-kDa peptide which originates from proteolysis of a larger protein precursor (APP) and accumulates in senile plaques in brains of Alzheimer's disease (AD) patients. Since secreted APP inhibits factors IXa, Xa and XIa, and thrombin appears to play a role in APP secretion and proteolysis, a relationship between hemostasis system and APP metabolism seems to exist. In this work we investigate the susceptibility to proteolytic cleavage by factor Xa of a fusion construct containing full-length APP prepared in bacteria, and demonstrate that both APP695 and APP770 are substrates for this protease. Factor Xa was found to cleave APP after arginines 102, 268, 510, 573 and 601 (APP695 numeration); most of these sites appear to be common for different coagulation factors. In addition, APP incubation with factor Xa generates an array of six potentially amyloidogenic fragments. Comparative kinetic analysis of APP695 and APP770 cleavage by factor Xa suggests that Kunitz-type inhibitor-containing isoforms exert an inhibitory effect on the protease. However, this inhibition is far from complete even at a 5-fold molar excess of inhibitor. Our results raise the possibility that proteases from the coagulation cascade may contribute to APP proteolysis, and support the notion that these proteases play a role in AD pathogenesis.
Collapse
Affiliation(s)
- C Haas
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Proteolytic processing of beta-amyloid precursor protein (APP) is believed to be fundamental to the understanding of Alzheimer's disease. The identities and the regulatory elements of the proteases involved in the process, known as alpha/beta/gamma secretases, are unclear. In this study, by examining reported data, we found some indications suggesting that the putative alpha-secretase may be a calcium-dependent protease, and that this enzyme may play a primary role in the regulation of APP processing. Based on this, we proposed a model for the membrane orientations of the secretases for further discussions.
Collapse
Affiliation(s)
- M Chen
- Department of Pharmacology and Therapeutics, University of South Florida College of Medicine, Tampa 33612, USA.
| |
Collapse
|
49
|
Abstract
Since the identification in 1984 of the amyloid beta protein (Abeta) as the major component of senile plaques and cerebrovascular amyloid in Alzheimer's disease (AD) brains, it is well accepted that the production of this protein is a crucial factor in the pathogenesis of AD. Abeta is produced by cleavage from the amyloid precursor protein (APP) and can form fibrils in vivo and in vitro. The formation of these fibrils is influenced by proteins that are found in association with Abeta-containing lesions in the AD brain. Several of these proteins arise by an inflammatory response of the brain to Abeta production. The distribution of different isoforms of Abeta, varying at the C-terminus of the peptide, varies among the Abeta-containing lesions in AD brains. Such variations may have consequences for the pathogenesis of AD because the various Abeta isoforms differ in their capacity to form fibrils, and they have different toxic effects on neurons and vascular cells, respectively. The experimental data indicate that the pathogenesis of senile plaques is different from the generation of cerebrovascular amyloidosis. Summarizing models for either type of AD pathology are presented.
Collapse
Affiliation(s)
- M M Verbeek
- Department of Pathology, University Hospital Nijmegen, The Netherlands
| | | | | |
Collapse
|
50
|
Salinero O, Moreno-Flores M, Ceballos M, Wandosell F. ?-Amyloid peptide induced cytoskeletal reorganization in cultured astrocytes. J Neurosci Res 1997. [DOI: 10.1002/(sici)1097-4547(19970115)47:2<216::aid-jnr10>3.0.co;2-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|