51
|
Cellular membrane fluidity in amyloid precursor protein processing. Mol Neurobiol 2014; 50:119-29. [PMID: 24553856 DOI: 10.1007/s12035-014-8652-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 01/23/2014] [Indexed: 12/20/2022]
Abstract
The senile plaque is a pathologic hallmark of Alzheimer's disease (AD). Amyloid-β peptide (Aβ), the main constituent of senile plaques, is neurotoxic especially in its oligomeric form. Aβ is derived from the sequential cleavage of amyloid precursor protein (APP) by β- and γ-secretases in the amyloidogenic pathway. Alternatively, APP can be cleaved by α-secretases within the Aβ domain to produce neurotrophic and neuroprotective α-secretase-cleaved soluble APP (sAPPα) in the nonamyloidogenic pathway. Since APP and α-, β-, and γ-secretases are membrane proteins, APP processing should be highly dependent on the membrane composition and the biophysical properties of cellular membrane. In this review, we discuss the role of the biophysical properties of cellular membrane in APP processing, especially the effects of phospholipases A(2) (PLA(2)s), fatty acids, cholesterol, and Aβ on membrane fluidity in relation to their effects on APP processing.
Collapse
|
52
|
Drolle E, Hane F, Lee B, Leonenko Z. Atomic force microscopy to study molecular mechanisms of amyloid fibril formation and toxicity in Alzheimer's disease. Drug Metab Rev 2014; 46:207-23. [PMID: 24495298 DOI: 10.3109/03602532.2014.882354] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by dementia and memory loss for which no cure or effective prevention is currently available. Neurodegeneration in AD is linked to formation of amyloid plaques found in brain tissues of Alzheimer's patients during post-mortem examination. Amyloid plaques are composed of amyloid fibrils and small oligomers - insoluble protein aggregates. Although amyloid plaques are found on the neuronal cell surfaces, the mechanism of amyloid toxicity is still not well understood. Currently, it is believed that the cytotoxicity is a result of the nonspecific interaction of small soluble amyloid oligomers (rather than longer fibrils) with the plasma membrane. In recent years, nanotechnology has contributed significantly to understanding the structure and function of lipid membranes and to the study of the molecular mechanisms of membrane-associated diseases. We review the current state of research, including applications of the latest nanotechnology approaches, on the interaction of lipid membranes with the amyloid-β (Aβ) peptide in relation to amyloid toxicity. We discuss the interactions of Aβ with model lipid membranes with a focus to demonstrate that composition, charge and phase of the lipid membrane, as well as lipid domains and rafts, affect the binding of Aβ to the membrane and contribute to toxicity. Understanding the role of the lipid membrane in AD at the nanoscale and molecular level will contribute to the understanding of the molecular mechanism of amyloid toxicity and may aid into the development of novel preventive strategies to combat AD.
Collapse
Affiliation(s)
- Elizabeth Drolle
- Department of Biology, University of Waterloo , Waterloo, ON , Canada
| | | | | | | |
Collapse
|
53
|
Saman S, Lee NC, Inoyo I, Jin J, Li Z, Doyle T, McKee AC, Hall GF. Proteins recruited to exosomes by tau overexpression implicate novel cellular mechanisms linking tau secretion with Alzheimer's disease. J Alzheimers Dis 2014; 40 Suppl 1:S47-70. [PMID: 24718102 PMCID: PMC5977388 DOI: 10.3233/jad-132135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Tau misprocessing to form aggregates and other toxic species has emerged as a major feature in our developing understanding of the etiology and pathogenesis of Alzheimer's disease (AD). The significance of tau misprocessing in AD has been further emphasized by recent studies showing that tau can be secreted from neurons via exosomes and may itself be an important agent in the spreading of neurofibrillary lesions within the brain. Tau secretion occurs most readily under disease-associated conditions in cellular models, suggesting that cellular changes responsible for secretion, possibly including tau oligomerization, could play a key role in the propagation of neurofibrillary lesions in neurodegenerative disease. Here we show that overexpression of 4R0N human tau in neuroblastoma cells recruits mitochondrial and axonogenesis-associated proteins relevant to neurodegeneration into the exosomal secretion pathway via distinct mechanisms. The recruitment of mitochondrial proteins appears to be linked to autophagy disruption (exophagy) in multiple neurodegenerative conditions but has few known direct links to AD and tau. By contrast, the involvement of synaptic plasticity and axonogenesis markers is highly specific to both tau and AD and may be relevant to the reactivation of developmental programs involving tau in AD and the recently demonstrated ability of secreted tau to establish tissue distribution gradients in CNS neuropil. We also found a highly significant correlation between genes that are significantly downregulated in multiple forms of AD and proteins that have been recruited to exosomes by tau, which we interpret as strong evidence for the central involvement of tau secretion in AD cytopathogenesis. Our results suggest that multiple cellular mechanisms may link tau secretion to both toxicity and neurofibrillary lesion spreading in AD and other tauopathies.
Collapse
Affiliation(s)
- Sudad Saman
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Street Lowell MA, USA
- Mass Bay Community College Science Department STEM Division 50 Oakland Street Wellesley Hills, MA 02481
| | - Norman C.Y. Lee
- Boston University Chemical Instrumentation Center, Department of Chemistry, Boston, MA USA 02215
| | - Itoro Inoyo
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Street Lowell MA, USA
| | - Jun Jin
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Street Lowell MA, USA
| | - Zhihan Li
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Street Lowell MA, USA
| | - Thomas Doyle
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Street Lowell MA, USA
- Mass Bay Community College Science Department STEM Division 50 Oakland Street Wellesley Hills, MA 02481
| | - Ann C. McKee
- GRECC unit, Veterans Administration Medical Center, 182-B, 200 Springs Rd, Bedford, MA 01730 and Departments of Neurology and Pathology, Boston University School of Medicine Boston, MA USA 02215
| | - Garth F. Hall
- Department of Biological Sciences, University of Massachusetts Lowell, 198 Riverside Street Lowell MA, USA
| |
Collapse
|
54
|
Membrane lipid modifications and therapeutic effects mediated by hydroxydocosahexaenoic acid on Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1680-92. [PMID: 24374316 DOI: 10.1016/j.bbamem.2013.12.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/16/2013] [Accepted: 12/18/2013] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative pathology with relevant unmet therapeutic needs. Both natural aging and AD have been associated with a significant decline in the omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA), and accordingly, administration of DHA has been proposed as a possible treatment for this pathology. However, recent clinical trials in mild-to-moderately affected patients have been inconclusive regarding the real efficacy of DHA in halting this disease. Here, we show that the novel hydroxyl-derivative of DHA (2-hydroxydocosahexaenoic acid - OHDHA) has a strong therapeutic potential to treat AD. We demonstrate that OHDHA administration increases DHA levels in the brain of a transgenic mouse model of AD (5xFAD), as well as those of phosphatidylethanolamine (PE) species that carry long polyunsaturated fatty acids (PUFAs). In 5xFAD mice, administration of OHDHA induced lipid modifications that were paralleled with a reduction in amyloid-β (Αβ) accumulation and full recovery of cognitive scores. OHDHA administration also reduced Aβ levels in cellular models of AD, in association with alterations in the subcellular distribution of secretases and reduced Aβ-induced tau protein phosphorylation as well. Furthermore, OHDHA enhanced the survival of neuron-like differentiated cells exposed to different insults, such as oligomeric Aβ and NMDA-mediated neurotoxicity. These results were supported by model membrane studies in which incorporation of OHDHA into lipid-raft-like vesicles was shown to reduce the binding affinity of oligomeric and fibrillar Aβ to membranes. Finally, the OHDHA concentrations used here did not produce relevant toxicity in zebrafish embryos in vivo. In conclusion, we demonstrate the pleitropic effects of OHDHA that might prove beneficial to treat AD, which suggests that an upstream event, probably the modulation of the membrane lipid composition and structure, influences cellular homeostasis reversing the neurodegenerative process. This Article is Part of a Special Issue Entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
|
55
|
Zempel H, Luedtke J, Kumar Y, Biernat J, Dawson H, Mandelkow E, Mandelkow EM. Amyloid-β oligomers induce synaptic damage via Tau-dependent microtubule severing by TTLL6 and spastin. EMBO J 2013; 32:2920-37. [PMID: 24065130 PMCID: PMC3831312 DOI: 10.1038/emboj.2013.207] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/22/2013] [Indexed: 01/23/2023] Open
Abstract
Mislocalization and aggregation of Aβ and Tau combined with loss of synapses and microtubules (MTs) are hallmarks of Alzheimer disease. We exposed mature primary neurons to Aβ oligomers and analysed changes in the Tau/MT system. MT breakdown occurs in dendrites invaded by Tau (Tau missorting) and is mediated by spastin, an MT-severing enzyme. Spastin is recruited by MT polyglutamylation, induced by Tau missorting triggered translocalization of TTLL6 (Tubulin-Tyrosine-Ligase-Like-6) into dendrites. Consequences are spine loss and mitochondria and neurofilament mislocalization. Missorted Tau is not axonally derived, as shown by axonal retention of photoconvertible Dendra2-Tau, but newly synthesized. Recovery from Aβ insult occurs after Aβ oligomers lose their toxicity and requires the kinase MARK (Microtubule-Affinity-Regulating-Kinase). In neurons derived from Tau-knockout mice, MTs and synapses are resistant to Aβ toxicity because TTLL6 mislocalization and MT polyglutamylation are prevented; hence no spastin recruitment and no MT breakdown occur, enabling faster recovery. Reintroduction of Tau re-establishes Aβ-induced toxicity in TauKO neurons, which requires phosphorylation of Tau's KXGS motifs. Transgenic mice overexpressing Tau show TTLL6 translocalization into dendrites and decreased MT stability. The results provide a rationale for MT stabilization as a therapeutic approach.
Collapse
Affiliation(s)
- Hans Zempel
- DZNE, German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
56
|
Chen RJ, Chang WW, Lin YC, Cheng PL, Chen YR. Alzheimer's amyloid-β oligomers rescue cellular prion protein induced tau reduction via the Fyn pathway. ACS Chem Neurosci 2013; 4:1287-96. [PMID: 23805846 DOI: 10.1021/cn400085q] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Amyloid-β (Aβ) and tau are the pathogenic hallmarks in Alzheimer's disease (AD). Aβ oligomers are considered the actual toxic entities, and the toxicity relies on the presence of tau. Recently, Aβ oligomers have been shown to specifically interact with cellular prion protein (PrP(C)) where the role of PrP(C) in AD is still not fully understood. To investigate the downstream mechanism of PrP(C) and Aβ oligomer interaction and their possible relationships to tau, we examined tau expression in human neuroblastoma BE(2)-C cells transfected with murine PrP(C) and studied the effect under Aβ oligomer treatment. By Western blotting, we found that PrP(C) overexpression down-regulated tau protein and Aβ oligomer binding alleviated the tau reduction induced by wild type but not M128V PrP(C), the high AD risk polymorphic allele in human prion gene. PrP(C) lacking the Aβ oligomer binding site was incapable of rescuing the level of tau reduction. Quantitative RT-PCR showed the PrP(C) effect was attributed to tau reduction at the transcription level. Treatment with Fyn pathway inhibitors, Fyn kinase inhibitor PP2 and MEK inhibitor U0126, reversed the PrP(C)-induced tau reduction and Aβ oligomer treatment modulated Fyn kinase activity. The results suggested Fyn pathway regulated Aβ-PrP(C)-tau signaling. Overall, our results demonstrated that PrP(C) down-regulated tau via the Fyn pathway and the effect can be regulated by Aβ oligomers. Our study facilitated the understanding of molecular mechanisms among PrP(C), tau, and Aβ oligomers.
Collapse
Affiliation(s)
- Rong-Jie Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Wei Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chun Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Pei-Lin Cheng
- Institute
of Molecular Biology, Academia Sinica,
Taipei, Taiwan
| | - Yun-Ru Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
57
|
Johnson RD, Schauerte JA, Chang CC, Wisser KC, Althaus JC, Carruthers CJL, Sutton MA, Steel DG, Gafni A. Single-molecule imaging reveals aβ42:aβ40 ratio-dependent oligomer growth on neuronal processes. Biophys J 2013; 104:894-903. [PMID: 23442968 DOI: 10.1016/j.bpj.2012.12.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 12/01/2012] [Accepted: 12/18/2012] [Indexed: 12/21/2022] Open
Abstract
Soluble oligomers of the amyloid-β peptide have been implicated as proximal neurotoxins in Alzheimer's disease. However, the identity of the neurotoxic aggregate(s) and the mechanisms by which these species induce neuronal dysfunction remain uncertain. Physiologically relevant experimentation is hindered by the low endogenous concentrations of the peptide, the metastability of Aβ oligomers, and the wide range of observed interactions between Aβ and biological membranes. Single-molecule microscopy represents one avenue for overcoming these challenges. Using this technique, we find that Aβ binds to primary rat hippocampal neurons at physiological concentrations. Although amyloid-β(1-40) as well as amyloid-β(1-42) initially form larger oligomers on neurites than on glass slides, a 1:1 mix of the two peptides result in smaller neurite-bound oligomers than those detected on-slide or for either peptide alone. With 1 nM peptide in solution, Aβ40 oligomers do not grow over the course of 48 h, Aβ42 oligomers grow slightly, and oligomers of a 1:1 mix grow substantially. Evidently, small Aβ oligomers are capable of binding to neurons at physiological concentrations and grow at rates dependent on local Aβ42:Aβ40 ratios. These results are intriguing in light of the increased Aβ42:Aβ40 ratios shown to correlate with familial Alzheimer's disease mutations.
Collapse
Affiliation(s)
- Robin D Johnson
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med 2013; 2:a006247. [PMID: 22762014 DOI: 10.1101/cshperspect.a006247] [Citation(s) in RCA: 572] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tau represents the subunit protein of one of the major hallmarks of Alzheimer disease (AD), the neurofibrillary tangles, and is therefore of major interest as an indicator of disease mechanisms. Many of the unusual properties of Tau can be explained by its nature as a natively unfolded protein. Examples are the large number of structural conformations and biochemical modifications (phosphorylation, proteolysis, glycosylation, and others), the multitude of interaction partners (mainly microtubules, but also other cytoskeletal proteins, kinases, and phosphatases, motor proteins, chaperones, and membrane proteins). The pathological aggregation of Tau is counterintuitive, given its high solubility, but can be rationalized by short hydrophobic motifs forming β structures. The aggregation of Tau is toxic in cell and animal models, but can be reversed by suppressing expression or by aggregation inhibitors. This review summarizes some of the structural, biochemical, and cell biological properties of Tau and Tau fibers. Further aspects of Tau as a diagnostic marker and therapeutic target, its involvement in other Tau-based diseases, and its histopathology are covered by other chapters in this volume.
Collapse
Affiliation(s)
- Eva-Maria Mandelkow
- Max-Planck Unit for Structural Molecular Biology, c/o DESY, 22607 Hamburg, Germany; DZNE, German Center for Neurodegenerative Diseases, and CAESAR Research Center, 53175 Bonn, Germany.
| | | |
Collapse
|
59
|
Noble W, Hanger DP, Miller CCJ, Lovestone S. The importance of tau phosphorylation for neurodegenerative diseases. Front Neurol 2013; 4:83. [PMID: 23847585 PMCID: PMC3696910 DOI: 10.3389/fneur.2013.00083] [Citation(s) in RCA: 313] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/14/2013] [Indexed: 01/20/2023] Open
Abstract
Fibrillar deposits of highly phosphorylated tau are a key pathological feature of several neurodegenerative tauopathies including Alzheimer's disease (AD) and some frontotemporal dementias. Increasing evidence suggests that the presence of these end-stage neurofibrillary lesions do not cause neuronal loss, but rather that alterations to soluble tau proteins induce neurodegeneration. In particular, aberrant tau phosphorylation is acknowledged to be a key disease process, influencing tau structure, distribution, and function in neurons. Although typically described as a cytosolic protein that associates with microtubules and regulates axonal transport, several additional functions of tau have recently been demonstrated, including roles in DNA stabilization, and synaptic function. Most recently, studies examining the trans-synaptic spread of tau pathology in disease models have suggested a potential role for extracellular tau in cell signaling pathways intrinsic to neurodegeneration. Here we review the evidence showing that tau phosphorylation plays a key role in neurodegenerative tauopathies. We also comment on the tractability of altering phosphorylation-dependent tau functions for therapeutic intervention in AD and related disorders.
Collapse
Affiliation(s)
- Wendy Noble
- Department of Neuroscience, King's College London, Institute of Psychiatry , London , UK
| | | | | | | |
Collapse
|
60
|
Ramachandran G, Udgaonkar JB. Mechanistic studies unravel the complexity inherent in tau aggregation leading to Alzheimer's disease and the tauopathies. Biochemistry 2013; 52:4107-26. [PMID: 23721410 DOI: 10.1021/bi400209z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aggregation of the protein tau into amyloid fibrils is known to be involved in the causation of the neurodegenerative tauopathies and the progression of cognitive decline in Alzheimer's disease. This review surveys the mechanism of tau aggregation with special emphasis on the information obtained from biochemical and biophysical studies. First, tau is described from a structure-function perspective. Subsequently, the connection of tau to neurodegeneration is explained, and a description of the tau amyloid fibril is provided. Lastly, studies of the mechanism of tau fibril formation are reviewed, and the physiological significance of these studies with reference to how they can clarify many aspects of disease progression is described. The aim of this review is to underscore how mechanistic studies reveal the complexity of the tau fibril formation pathway and the plethora of species populated on or off the pathway of aggregation, and how this information can be beneficial in the design of inhibitors or drugs that ameliorate neurodegeneration.
Collapse
Affiliation(s)
- Gayathri Ramachandran
- National Centre for Biological Sciences, Tata Institute of Fundamental Research , Bangalore 560065, India
| | | |
Collapse
|
61
|
Lipid raft disarrangement as a result of neuropathological progresses: a novel strategy for early diagnosis? Neuroscience 2013; 245:26-39. [PMID: 23618758 DOI: 10.1016/j.neuroscience.2013.04.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/06/2013] [Accepted: 04/08/2013] [Indexed: 11/21/2022]
Abstract
Lipid rafts are the preferential site of numerous membrane signaling proteins which are involved in neuronal functioning and survival. These proteins are organized in multiprotein complexes, or signalosomes, in close contact with lipid classes particularly represented in lipid rafts (i.e. cholesterol, sphingolipids and saturated fatty acids), which may contribute to physiological responses leading to neuroprotection. Increasing evidence indicates that alteration of lipid composition in raft structures as a consequence of neuropathologies, such as Alzheimer's disease (AD) and Parkinson's disease (PD), causes a dramatic increase in lipid raft order. These phenomena may correlate with perturbation of signalosome activities, likely contributing to neurodegenerative progression. Interestingly, significant disruption of stable raft microenvironments has been already observed in the first stages of either AD or PD, suggesting that these alterations may represent early events in the neuropathological development. In this regard, the search for biochemical markers, such as specific metabolic products altered in the brain at the first steps of the disease, presently represents an important challenge for early diagnostic strategies. Alterations of these biomarkers may be reflected in either plasma or cerebrospinal fluid, thus representing a potential strategy to predict an accurate diagnosis. We propose that pathologically-linked lipid raft markers may be interesting candidates to be explored at this level, although it has not been studied so far to what extent alteration of different signalosome components may be reflected in peripheral fluids. In this mini-review, we will discuss on relevant aspects of lipid rafts that contribute to the modulation of neuropathological events related to AD and PD. An interesting hypothesis is that anomalies on raft biomarkers measured at peripheral fluids might mirror the lipid raft pathology observed in early stages of AD and PD.
Collapse
|
62
|
Partitioning and confinement of GM1 ganglioside induced by amyloid aggregates. FEBS Lett 2013; 587:1385-91. [DOI: 10.1016/j.febslet.2013.03.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 02/22/2013] [Accepted: 03/07/2013] [Indexed: 12/13/2022]
|
63
|
Burke KA, Yates EA, Legleiter J. Biophysical insights into how surfaces, including lipid membranes, modulate protein aggregation related to neurodegeneration. Front Neurol 2013; 4:17. [PMID: 23459674 PMCID: PMC3585431 DOI: 10.3389/fneur.2013.00017] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/09/2013] [Indexed: 11/13/2022] Open
Abstract
There are a vast number of neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD), associated with the rearrangement of specific proteins to non-native conformations that promotes aggregation and deposition within tissues and/or cellular compartments. These diseases are commonly classified as protein-misfolding or amyloid diseases. The interaction of these proteins with liquid/surface interfaces is a fundamental phenomenon with potential implications for protein-misfolding diseases. Kinetic and thermodynamic studies indicate that significant conformational changes can be induced in proteins encountering surfaces, which can play a critical role in nucleating aggregate formation or stabilizing specific aggregation states. Surfaces of particular interest in neurodegenerative diseases are cellular and subcellular membranes that are predominately comprised of lipid components. The two-dimensional liquid environments provided by lipid bilayers can profoundly alter protein structure and dynamics by both specific and non-specific interactions. Importantly for misfolding diseases, these bilayer properties can not only modulate protein conformation, but also exert influence on aggregation state. A detailed understanding of the influence of (sub)cellular surfaces in driving protein aggregation and/or stabilizing specific aggregate forms could provide new insights into toxic mechanisms associated with these diseases. Here, we review the influence of surfaces in driving and stabilizing protein aggregation with a specific emphasis on lipid membranes.
Collapse
Affiliation(s)
- Kathleen A Burke
- C. Eugene Bennett Department of Chemistry, West Virginia University Morgantown, WV, USA
| | | | | |
Collapse
|
64
|
Rushworth JV, Griffiths HH, Watt NT, Hooper NM. Prion protein-mediated toxicity of amyloid-β oligomers requires lipid rafts and the transmembrane LRP1. J Biol Chem 2013; 288:8935-51. [PMID: 23386614 PMCID: PMC3610967 DOI: 10.1074/jbc.m112.400358] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Soluble oligomers of the amyloid-β (Aβ) peptide cause neurotoxicity, synaptic dysfunction, and memory impairments that underlie Alzheimer disease (AD). The cellular prion protein (PrPC) was recently identified as a high affinity neuronal receptor for Aβ oligomers. We report that fibrillar Aβ oligomers recognized by the OC antibody, which have been shown to correlate with the onset and severity of AD, bind preferentially to cells and neurons expressing PrPC. The binding of Aβ oligomers to cell surface PrPC, as well as their downstream activation of Fyn kinase, was dependent on the integrity of cholesterol-rich lipid rafts. In SH-SY5Y cells, fluorescence microscopy and co-localization with subcellular markers revealed that the Aβ oligomers co-internalized with PrPC, accumulated in endosomes, and subsequently trafficked to lysosomes. The cell surface binding, internalization, and downstream toxicity of Aβ oligomers was dependent on the transmembrane low density lipoprotein receptor-related protein-1 (LRP1). The binding of Aβ oligomers to cell surface PrPC impaired its ability to inhibit the activity of the β-secretase BACE1, which cleaves the amyloid precursor protein to produce Aβ. The green tea polyphenol (−)-epigallocatechin gallate and the red wine extract resveratrol both remodeled the fibrillar conformation of Aβ oligomers. The resulting nonfibrillar oligomers displayed significantly reduced binding to PrPC-expressing cells and were no longer cytotoxic. These data indicate that soluble, fibrillar Aβ oligomers bind to PrPC in a conformation-dependent manner and require the integrity of lipid rafts and the transmembrane LRP1 for their cytotoxicity, thus revealing potential targets to alleviate the neurotoxic properties of Aβ oligomers in AD.
Collapse
Affiliation(s)
- Jo V Rushworth
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | | | | | | |
Collapse
|
65
|
Abstract
There is growing evidence that cell membranes can contain domains with different lipid and protein compositions and with different physical properties. Furthermore, it is increasingly appreciated that sphingolipids play a crucial role in the formation and properties of ordered lipid domains (rafts) in cell membranes. This review describes recent advances in our understanding of ordered membrane domains in both cells and model membranes. In addition, how the structure of sphingolipids influences their ability to participate in the formation of ordered domains, as well as how sphingolipid structure alters ordered domain properties, is described. The diversity of sphingolipid structure is likely to play an important role in modulating the biologically relevant properties of "rafts" in cell membranes.
Collapse
|
66
|
Larson M, Sherman MA, Amar F, Nuvolone M, Schneider JA, Bennett DA, Aguzzi A, Lesné SE. The complex PrP(c)-Fyn couples human oligomeric Aβ with pathological tau changes in Alzheimer's disease. J Neurosci 2012; 32:16857-71a. [PMID: 23175838 PMCID: PMC3568961 DOI: 10.1523/jneurosci.1858-12.2012] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 09/21/2012] [Accepted: 09/25/2012] [Indexed: 01/01/2023] Open
Abstract
Amid controversy, the cellular form of the prion protein PrP(c) has been proposed to mediate oligomeric amyloid-β (Aβ)-induced deficits. In contrast, there is consistent evidence that the Src kinase Fyn is activated by Aβ oligomers and leads to synaptic and cognitive impairment in transgenic animals. However, the molecular mechanism by which soluble Aβ activates Fyn remains unknown. Combining the use of human and transgenic mouse brain tissue as well as primary cortical neurons, we demonstrate that soluble Aβ binds to PrP(c) at neuronal dendritic spines in vivo and in vitro where it forms a complex with Fyn, resulting in the activation of the kinase. Using the antibody 6D11 to prevent oligomeric Aβ from binding to PrP(c), we abolished Fyn activation and Fyn-dependent tau hyperphosphorylation induced by endogenous oligomeric Aβ in vitro. Finally, we showed that gene dosage of Prnp regulates Aβ-induced Fyn/tau alterations. Together, our findings identify a complete signaling cascade linking one specific endogenous Aβ oligomer, Fyn alteration, and tau hyperphosphorylation in cellular and animal models modeling aspects of the molecular pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Megan Larson
- Departments of Neuroscience
- N. Bud Grossman Center for Memory Research and Care, and
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55414
| | - Mathew A. Sherman
- Departments of Neuroscience
- N. Bud Grossman Center for Memory Research and Care, and
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55414
| | - Fatou Amar
- Departments of Neuroscience
- N. Bud Grossman Center for Memory Research and Care, and
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55414
| | - Mario Nuvolone
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
- Amyloid Center, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Department of Molecular Medicine, University of Pavia and Istituto Universitario di Studi Superiori di Pavia, I-27100 Pavia, Italy, and
| | - Julie A. Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Sylvain E. Lesné
- Departments of Neuroscience
- N. Bud Grossman Center for Memory Research and Care, and
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55414
| |
Collapse
|
67
|
Um JW, Nygaard HB, Heiss JK, Kostylev MA, Stagi M, Vortmeyer A, Wisniewski T, Gunther EC, Strittmatter SM. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci 2012; 15:1227-35. [PMID: 22820466 PMCID: PMC3431439 DOI: 10.1038/nn.3178] [Citation(s) in RCA: 520] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 07/02/2012] [Indexed: 12/21/2022]
Abstract
Amyloid-beta (Aβ) oligomers are thought to trigger Alzheimer’s disease (AD) pathophysiology. Cellular Prion Protein (PrPC) selectively binds oligomeric Aβ and can mediate AD-related phenotypes. Here, we examined the specificity, distribution and signaling from Aβ/PrP complexes, seeking to explain how they might alter the function of NMDA receptors in neurons. PrPC is enriched in post-synaptic densities, and Aβ/PrPC interaction leads to Fyn kinase activation. Soluble Aβ assemblies derived from human AD brain interact with PrPC to activate Fyn. Aβ engagement of PrPC/Fyn signaling yields phosphorylation of the NR2B subunit of NMDA-receptors, which is coupled to an initial increase and then loss of surface NMDA-receptors. Aβ-induced LDH release and dendritic spine loss require both PrPC and Fyn, and human familial AD transgene-induced convulsive seizures do not occur in mice lacking PrPC. These results delineate an Aβ oligomer signal transduction pathway requiring PrPC and Fyn to alter synaptic function with relevance to AD.
Collapse
Affiliation(s)
- Ji Won Um
- Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Wang J, Ohno-Matsui K, Morita I. Cholesterol enhances amyloid β deposition in mouse retina by modulating the activities of Aβ-regulating enzymes in retinal pigment epithelial cells. Biochem Biophys Res Commun 2012; 424:704-9. [PMID: 22796523 DOI: 10.1016/j.bbrc.2012.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/06/2012] [Indexed: 10/28/2022]
Abstract
Subretinally-deposited amyloid β (Aβ) is a main contributor of developing age-related macular degeneration (AMD). However, the mechanism causing Aβ deposition in AMD eyes is unknown. Hypercholesterolemia is a significant risk for developing AMD. Thus, we investigated the effects of cholesterol on Aβ production in retinal pigment epithelial (RPE) cells in vitro and in the mouse retina in vivo. RPE cells isolated from senescent (12-month-old) C57BL/6 mice were treated with 10μg/ml cholesterol for 48h. Aβ amounts in culture supernatants were measured by ELISA. Activity and expression of enzymes and proteins that regulate Aβ production were examined by activity assay and real time PCR. The retina of mice fed cholesterol-enriched diet was examined by transmission electron microscopy. Cholesterol significantly increased Aβ production in cultured RPE cells. Activities of Aβ degradation enzyme; neprilysin (NEP) and anti-amyloidogenic secretase; α-secretase were significantly decreased in cell lysates of cholesterol-treated RPE cells compared to non-treated cells, but there was no change in the activities of β- or γ-secretase. mRNA levels of NEP and α-secretase (ADAM10 and ADAM17) were significantly lower in cholesterol-treated RPE cells than non-treated cells. Senescent (12-month-old) mice fed cholesterol-enriched chow developed subRPE deposits containing Aβ, whereas age-matched mice fed standard rodent chow diet did not. Activities and mRNA levels of NEP and α-secretase were significantly lower in native RPE cells freshly isolated from cholesterol-enriched chow fed mice compared to standard rodent chow fed mice. These findings suggest that cholesterol enhances subretinal Aβ accumulation by modulating the activities of enzymes degrading and processing Aβ in RPE cells in senescent subjects.
Collapse
Affiliation(s)
- Jiying Wang
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | | |
Collapse
|
69
|
Mazargui H, Lévêque C, Bartnik D, Fantini J, Gouget T, Melone MAB, Funke SA, Willbold D, Perrone L. A synthetic amino acid substitution of Tyr10 in Aβ peptide sequence yields a dominant negative variant in amyloidogenesis. Aging Cell 2012; 11:530-41. [PMID: 22385841 DOI: 10.1111/j.1474-9726.2012.00814.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in elderly people, and age is the major nongenetic risk factor for sporadic AD. A hallmark of AD is the accumulation of amyloid in the brain, which is composed mainly of the amyloid beta-peptide (Aβ) in the form of oligomers and fibrils. However, how aging induces Aβ aggregation is not yet fully determined. Some residues in the Aβ sequence seem to promote Aβ-induced toxicity in association with age-dependent risk factors for AD, such as (i) increased GM1 brain membrane content, (ii) altered lipid domain in brain membrane, (iii) oxidative stress. However, the role of Aβ sequence in promoting aggregation following interaction with the plasma membrane is not yet demonstrated. As Tyr10 is implicated in the induction of oxidative stress and stabilization of Aβ aggregation, we substituted Tyr 10 with a synthetic amino acid that abolishes Aβ-induced oxidative stress and shows an accelerated interaction with GM1. This variant peptide shows impaired aggregation properties and increased affinity for GM1. It has a dominant negative effect on amyloidogenesis in vitro, in cellulo, and in isolated synaptosomes. The present study shed new light in the understanding of Aβ-membrane interactions in Aβ-induced neurotoxicity. It demonstrates the relevance of Aβ sequence in (i) Aβ-membrane interaction, underlining the role of age-dependent enhanced GM1 content in promoting Aβ aggregation, (ii) Aβ aggregation, and (iii) Aβ-induced oxidative stress. Our results open the way for the design of peptides aimed to inhibit Aβ aggregation and neurotoxicity.
Collapse
|
70
|
Oligomeric amyloid-β peptide affects the expression of genes involved in steroid and lipid metabolism in primary neurons. Neurochem Int 2012; 61:321-33. [PMID: 22579571 DOI: 10.1016/j.neuint.2012.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 04/10/2012] [Accepted: 05/01/2012] [Indexed: 01/28/2023]
Abstract
Amyloid-β peptide (Aβ) is the principal component of plaques in the brains of patients with Alzheimer's disease (AD), and the most toxic form of Aβ may be as soluble oligomers. We report here the results of a microarray study of gene expression profiles in primary mouse cortical neurons in response to oligomeric Aβ(1-42). A major and unexpected finding was the down-regulation of genes involved in the biosynthesis of cholesterol and other steroids and lipids (such as Fdft1, Fdps, Idi1, Ldr, Mvd, Mvk, Nsdhl, Sc4mol), the expression of which was verified by quantitative real-time RT-PCR (qPCR). The ATP-binding cassette gene Abca1, which has a major role in cholesterol transport in brain and other tissues and has been genetically linked to AD, was notably up-regulated. The possible involvement of cholesterol and other lipids in Aβ synthesis and action in Alzheimer's disease has been studied and debated extensively but remains unresolved. These new data suggest that Aβ may influence steroid and lipid metabolism in neurons via multiple gene-expression changes.
Collapse
|
71
|
Jones EM, Dubey M, Camp PJ, Vernon BC, Biernat J, Mandelkow E, Majewski J, Chi EY. Interaction of tau protein with model lipid membranes induces tau structural compaction and membrane disruption. Biochemistry 2012; 51:2539-50. [PMID: 22401494 DOI: 10.1021/bi201857v] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The misfolding and aggregation of the intrinsically disordered, microtubule-associated tau protein into neurofibrillary tangles is implicated in the pathogenesis of Alzheimer's disease. However, the mechanisms of tau aggregation and toxicity remain unknown. Recent work has shown that anionic lipid membranes can induce tau aggregation and that membrane permeabilization may serve as a pathway by which protein aggregates exert toxicity, suggesting that the plasma membrane may play dual roles in tau pathology. This prompted our investigation to assess tau's propensity to interact with membranes and to elucidate the mutually disruptive structural perturbations the interactions induce in both tau and the membrane. We show that although highly charged and soluble, the full-length tau (hTau40) is also highly surface active, selectively inserts into anionic DMPG lipid monolayers and induces membrane morphological changes. To resolve molecular-scale structural details of hTau40 associated with lipid membranes, X-ray and neutron scattering techniques are utilized. X-ray reflectivity indicates hTau40s presence underneath a DMPG monolayer and penetration into the lipid headgroups and tailgroups, whereas grazing incidence X-ray diffraction shows that hTau40 insertion disrupts lipid packing. Moreover, both air/water and DMPG lipid membrane interfaces induce the disordered hTau40 to partially adopt a more compact conformation with density similar to that of a folded protein. Neutron reflectivity shows that tau completely disrupts supported DMPG bilayers while leaving the neutral DPPC bilayer intact. Our results show that hTau40s strong interaction with anionic lipids induces tau structural compaction and membrane disruption, suggesting possible membrane-based mechanisms of tau aggregation and toxicity in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmalee M Jones
- Department of Chemical and Nuclear Engineering, Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Abstract
Tauopathies are age-related neurodegenerative diseases that are characterized by the presence of aggregates of abnormally phosphorylated tau. As tau was originally discovered as a microtubule-associated protein, it has been hypothesized that neurodegeneration results from a loss of the ability of tau to associate with microtubules. However, tau has been found to have other functions aside from the promotion and stabilization of microtubule assembly. It is conceivable that such functions may be affected by the abnormal phosphorylation of tau and might have consequences for neuronal function or viability. This chapter provides an overview of tau structure, functions, and its involvement in neurodegenerative diseases.
Collapse
|
73
|
Sapir T, Frotscher M, Levy T, Mandelkow EM, Reiner O. Tau's role in the developing brain: implications for intellectual disability. Hum Mol Genet 2011; 21:1681-92. [PMID: 22194194 DOI: 10.1093/hmg/ddr603] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Microdeletions encompassing the MAPT (Tau) locus resulting in intellectual disability raised the hypothesis that Tau may regulate early functions in the developing brain. Our results indicate that neuronal migration was inhibited in mouse brains following Tau reduction. In addition, the leading edge of radially migrating neurons was aberrant in spite of normal morphology of radial glia. Furthermore, intracellular mitochondrial transport and morphology were affected. In early postnatal brains, a portion of Tau knocked down neurons reached the cortical plate. Nevertheless, they exhibited far less developed dendrites and a striking reduction in connectivity evident by the size of boutons. Our novel results strongly implicate MAPT as a dosage-sensitive gene in this locus involved in intellectual disability. Furthermore, our results are likely to impact our understanding of other diseases involving Tau.
Collapse
Affiliation(s)
- Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | | | | | | | | |
Collapse
|
74
|
Bucciantini M, Nosi D, Forzan M, Russo E, Calamai M, Pieri L, Formigli L, Quercioli F, Soria S, Pavone F, Savistchenko J, Melki R, Stefani> M. Toxic effects of amyloid fibrils on cell membranes: the importance of ganglioside GM1. FASEB J 2011; 26:818-31. [DOI: 10.1096/fj.11-189381] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Monica Bucciantini
- Department of Biochemical Sciences, and Forensic MedicineUniversity of Florence Florence Italy
- Research Centre on the Molecular Basis of Neurodegeneration, and Forensic MedicineUniversity of Florence Florence Italy
| | - Daniele Nosi
- Department of Anatomy, Histology, and Forensic MedicineUniversity of Florence Florence Italy
| | - Mario Forzan
- Department of Animal Pathology, Food Prophylaxis, and HygieneUniversity of Pisa Pisa Italy
| | - Edda Russo
- Department of Biochemical Sciences, and Forensic MedicineUniversity of Florence Florence Italy
| | - Martino Calamai
- European Laboratory for Nonlinear Spectroscopy (LENS)University of Florence Florence Italy
| | - Laura Pieri
- Laboratoire d'Enzymologie et Biochimie StructuralesCentre National de la Recherche Scientifique Gif sur Yvette France
| | - Lucia Formigli
- Department of Anatomy, Histology, and Forensic MedicineUniversity of Florence Florence Italy
| | - Franco Quercioli
- National Institute of OpticsConsiglio Nazionale delle Ricerche Florence Research Area Florence Italy
| | - Silvia Soria
- Nello Carrara Institute of Applied PhysicsConsiglio Nazionale delle Ricerche Florence Research Area Florence Italy
| | - Francesco Pavone
- European Laboratory for Nonlinear Spectroscopy (LENS)University of Florence Florence Italy
| | - Jimmy Savistchenko
- Laboratoire d'Enzymologie et Biochimie StructuralesCentre National de la Recherche Scientifique Gif sur Yvette France
| | - Ronald Melki
- Department of Animal Pathology, Food Prophylaxis, and HygieneUniversity of Pisa Pisa Italy
- Laboratoire d'Enzymologie et Biochimie StructuralesCentre National de la Recherche Scientifique Gif sur Yvette France
| | - Massimo Stefani>
- Department of Biochemical Sciences, and Forensic MedicineUniversity of Florence Florence Italy
- Research Centre on the Molecular Basis of Neurodegeneration, and Forensic MedicineUniversity of Florence Florence Italy
| |
Collapse
|
75
|
Legleiter J, Fryer JD, Holtzman DM, Kowalewski T. The modulating effect of mechanical changes in lipid bilayers caused by apoE-containing lipoproteins on Aβ induced membrane disruption. ACS Chem Neurosci 2011; 2:588-599. [PMID: 22125665 DOI: 10.1021/cn2000475] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A major feature of Alzheimer's disease (AD), a late-onset neurodegenerative disorder, is the ordered aggregation of the β-amyloid peptide (Aβ) into fibrils that comprise extracellular neuritic plaques found in the disease brain. One of many potential pathways for Aβ toxicity may be modulation of lipid membrane function. Here, we show by in situ atomic force microscopy (AFM) that astrocyte secreted lipoprotein particles (ASLPs) containing different isoforms of apolipoprotein E (apoE), of which the apoE4 allele is a major risk factor for the development of AD, can protect total brain lipid extract bilayers from Aβ(1-40) induced disruption. The apoE4 allele was less effective in protecting lipid bilayers from disruption compared with apoE3. Size analysis of apoE-containing ASLPs and mechanical studies of bilayer properties revealed that apoE-containing ASLPs modulate the mechanical properties of bilayers by acquiring some bilayer components (most likely cholesterol and/or oxidatively damaged lipids). Measurement of bilayer mechanical properties was accomplished with scanning probe acceleration microscopy (SPAM). These measurements demonstrated that apoE4 was also less effective in modulating mechanical properties of bilayers in comparison with apoE3. This ability of apoE to alter the mechanical properties of lipid membranes may represent a potential mechanism for the suppression of Aβ(1-40) induced bilayer disruption.
Collapse
Affiliation(s)
- Justin Legleiter
- The C. Eugene Bennett Department of Chemistry, WVnano Initiative, the Center for Neurosciences, West Virginia University, 217 Clark Hall, P.O. Box 6045, Morgantown, West Virginia 26506, United States
| | - John D. Fryer
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St. Louis, Missouri 63110, United States
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Tomasz Kowalewski
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
76
|
Johnson RD, Schauerte JA, Wisser KC, Gafni A, Steel DG. Direct observation of single amyloid-β(1-40) oligomers on live cells: binding and growth at physiological concentrations. PLoS One 2011; 6:e23970. [PMID: 21901146 PMCID: PMC3162019 DOI: 10.1371/journal.pone.0023970] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 07/28/2011] [Indexed: 11/18/2022] Open
Abstract
Understanding how amyloid-β peptide interacts with living cells on a molecular level is critical to development of targeted treatments for Alzheimer's disease. Evidence that oligomeric Aβ interacts with neuronal cell membranes has been provided, but the mechanism by which membrane binding occurs and the exact stoichiometry of the neurotoxic aggregates remain elusive. Physiologically relevant experimentation is hindered by the high Aβ concentrations required for most biochemical analyses, the metastable nature of Aβ aggregates, and the complex variety of Aβ species present under physiological conditions. Here we use single molecule microscopy to overcome these challenges, presenting direct optical evidence that small Aβ(1-40) oligomers bind to living neuroblastoma cells at physiological Aβ concentrations. Single particle fluorescence intensity measurements indicate that cell-bound Aβ species range in size from monomers to hexamers and greater, with the majority of bound oligomers falling in the dimer-to-tetramer range. Furthermore, while low-molecular weight oligomeric species do form in solution, the membrane-bound oligomer size distribution is shifted towards larger aggregates, indicating either that bound Aβ oligomers can rapidly increase in size or that these oligomers cluster at specific sites on the membrane. Calcium indicator studies demonstrate that small oligomer binding at physiological concentrations induces only mild, sporadic calcium leakage. These findings support the hypothesis that small oligomers are the primary Aβ species that interact with neurons at physiological concentrations.
Collapse
Affiliation(s)
- Robin D Johnson
- Department of Biophysics, The University of Michigan, Ann Arbor, Michigan, United States of America.
| | | | | | | | | |
Collapse
|
77
|
Usardi A, Pooler AM, Seereeram A, Reynolds CH, Derkinderen P, Anderton B, Hanger DP, Noble W, Williamson R. Tyrosine phosphorylation of tau regulates its interactions with Fyn SH2 domains, but not SH3 domains, altering the cellular localization of tau. FEBS J 2011; 278:2927-37. [PMID: 21692989 DOI: 10.1111/j.1742-4658.2011.08218.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent reports have demonstrated that interactions between the microtubule-associated protein tau and the nonreceptor tyrosine kinase Fyn play a critical role in mediating synaptic toxicity and neuronal loss in response to β-amyloid (Aβ) in models of Alzheimer's disease. Disruption of interactions between Fyn and tau may thus have the potential to protect neurons from Aβ-induced neurotoxicity. Here, we investigated tau and Fyn interactions and the potential implications for positioning of these proteins in membrane microdomains. Tau is known to bind to Fyn via its Src-homology (SH)3 domain, an association regulated by phosphorylation of PXXP motifs in tau. Here, we show that Pro216 within the PXXP(213-216) motif in tau plays an important role in mediating the interaction of tau with Fyn-SH3. We also show that tau interacts with the SH2 domain of Fyn, and that this association, unlike that of Fyn-SH3, is influenced by Fyn-mediated tyrosine phosphorylation of tau. In particular, phosphorylation of tau at Tyr18, a reported target of Fyn, is important for mediating Fyn-SH2-tau interactions. Finally, we show that tyrosine phosphorylation influences the localization of tau to detergent-resistant membrane microdomains in primary cortical neurons, and that this trafficking is Fyn-dependent. These findings may have implications for the development of novel therapeutic strategies aimed at disrupting the tau/Fyn-mediated synaptic dysfunction that occurs in response to elevated Aβ levels in neurodegenerative disease.
Collapse
Affiliation(s)
- Alessia Usardi
- Department of Neuroscience, MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Kawahara M, Ohtsuka I, Yokoyama S, Kato-Negishi M, Sadakane Y. Membrane Incorporation, Channel Formation, and Disruption of Calcium Homeostasis by Alzheimer's β-Amyloid Protein. Int J Alzheimers Dis 2011; 2011:304583. [PMID: 21547225 PMCID: PMC3087492 DOI: 10.4061/2011/304583] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 12/22/2010] [Accepted: 01/27/2011] [Indexed: 01/09/2023] Open
Abstract
Oligomerization, conformational changes, and the consequent neurodegeneration of Alzheimer's β-amyloid protein (AβP) play crucial roles in the pathogenesis of Alzheimer's disease (AD). Mounting evidence suggests that oligomeric AβPs cause the disruption of calcium homeostasis, eventually leading to neuronal death. We have demonstrated that oligomeric AβPs directly incorporate into neuronal membranes, form cation-sensitive ion channels (“amyloid channels”), and cause the disruption of calcium homeostasis via the amyloid channels. Other disease-related amyloidogenic proteins, such as prion protein in prion diseases or α-synuclein in dementia with Lewy bodies, exhibit similarities in the incorporation into membranes and the formation of calcium-permeable channels. Here, based on our experimental results and those of numerous other studies, we review the current understanding of the direct binding of AβP into membrane surfaces and the formation of calcium-permeable channels. The implication of composition of membrane lipids and the possible development of new drugs by influencing membrane properties and attenuating amyloid channels for the treatment and prevention of AD is also discussed.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Analytical Chemistry, School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, 1714-1 Yoshino-cho, Nobeoka-shi, Miyazaki 882-8508, Japan
| | | | | | | | | |
Collapse
|
79
|
De Vos A, Anandhakumar J, Van den Brande J, Verduyckt M, Franssens V, Winderickx J, Swinnen E. Yeast as a model system to study tau biology. Int J Alzheimers Dis 2011; 2011:428970. [PMID: 21559193 PMCID: PMC3090044 DOI: 10.4061/2011/428970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 01/21/2011] [Indexed: 11/20/2022] Open
Abstract
Hyperphosphorylated and aggregated human protein tau constitutes a hallmark of a multitude of neurodegenerative diseases called tauopathies, exemplified by Alzheimer's disease. In spite of an enormous amount of research performed on tau biology, several crucial questions concerning the mechanisms of tau toxicity remain unanswered. In this paper we will highlight some of the processes involved in tau biology and pathology, focusing on tau phosphorylation and the interplay with oxidative stress. In addition, we will introduce the development of a human tau-expressing yeast model, and discuss some crucial results obtained in this model, highlighting its potential in the elucidation of cellular processes leading to tau toxicity.
Collapse
Affiliation(s)
- Ann De Vos
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Jayamani Anandhakumar
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Jeff Van den Brande
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Mathias Verduyckt
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Vanessa Franssens
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Joris Winderickx
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Erwin Swinnen
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| |
Collapse
|
80
|
Askarova S, Yang X, Lee JCM. Impacts of membrane biophysics in Alzheimer's disease: from amyloid precursor protein processing to aβ Peptide-induced membrane changes. Int J Alzheimers Dis 2011; 2011:134971. [PMID: 21547213 PMCID: PMC3087431 DOI: 10.4061/2011/134971] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 12/30/2010] [Accepted: 01/21/2011] [Indexed: 12/11/2022] Open
Abstract
An increasing amount of evidence supports the notion that cytotoxic effects of amyloid-β peptide (Aβ), the main constituent of senile plaques in Alzheimer's disease (AD), are strongly associated with its ability to interact with membranes of neurons and other cerebral cells. Aβ is derived from amyloidogenic cleavage of amyloid precursor protein (AβPP) by β- and γ-secretase. In the nonamyloidogenic pathway, AβPP is cleaved by α-secretases. These two pathways compete with each other, and enhancing the non-amyloidogenic pathway has been suggested as a potential pharmacological approach for the treatment of AD. Since AβPP, α-, β-, and γ-secretases are membrane-associated proteins, AβPP processing and Aβ production can be affected by the membrane composition and properties. There is evidence that membrane composition and properties, in turn, play a critical role in Aβ cytotoxicity associated with its conformational changes and aggregation into oligomers and fibrils. Understanding the mechanisms leading to changes in a membrane's biophysical properties and how they affect AβPP processing and Aβ toxicity should prove to provide new therapeutic strategies for prevention and treatment of AD.
Collapse
Affiliation(s)
- Sholpan Askarova
- Department of Biological Engineering, University of Missouri, Columbia, MO 65211, USA
| | | | | |
Collapse
|
81
|
Pooler AM, Usardi A, Evans CJ, Philpott KL, Noble W, Hanger DP. Dynamic association of tau with neuronal membranes is regulated by phosphorylation. Neurobiol Aging 2011; 33:431.e27-38. [PMID: 21388709 DOI: 10.1016/j.neurobiolaging.2011.01.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 12/10/2010] [Accepted: 01/15/2011] [Indexed: 10/18/2022]
Abstract
Tau is an abundant cytosolic protein which regulates cytoskeletal stability by associating with microtubules in a phosphorylation-dependent manner. We have found a significant proportion of tau is located in the membrane fraction of rat cortical neurons and is dephosphorylated, at least at Tau-1 (Ser199/Ser202), AT8 (Ser199/Ser202/Thr205) and PHF-1 (Ser396/Ser404) epitopes. Inhibition of tau kinases casein kinase 1 (CK1) or glycogen synthase kinase-3 decreased tau phosphorylation and significantly increased amounts of tau in the membrane fraction. Mutation of serine/threonine residues to glutamate to mimic phosphorylation in the N-terminal, but not C-terminal, region of tau prevented its membrane localization in transfected cells, demonstrating that the phosphorylation state of tau directly impacts its localization. Inhibiting CK1 in neurons lacking the tyrosine kinase fyn also induced tau dephosphorylation but did not affect its membrane association. Furthermore, inhibition of CK1 increased binding of neuronal tau to the fyn-SH3 domain. We conclude that trafficking of tau between the cytosol and the neuronal membrane is dynamically regulated by tau phosphorylation through a mechanism dependent on fyn expression.
Collapse
Affiliation(s)
- Amy M Pooler
- King's College London, MRC Centre for Neurodegeneration Research, Department of Neuroscience, Institute of Psychiatry, London, UK.
| | | | | | | | | | | |
Collapse
|
82
|
Scales TM, Derkinderen P, Leung KY, Byers HL, Ward MA, Price C, Bird IN, Perera T, Kellie S, Williamson R, Anderton BH, Reynolds CH. Tyrosine phosphorylation of tau by the SRC family kinases lck and fyn. Mol Neurodegener 2011; 6:12. [PMID: 21269457 PMCID: PMC3037338 DOI: 10.1186/1750-1326-6-12] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 01/26/2011] [Indexed: 11/10/2022] Open
Abstract
Background Tau protein is the principal component of the neurofibrillary tangles found in Alzheimer's disease, where it is hyperphosphorylated on serine and threonine residues, and recently phosphotyrosine has been demonstrated. The Src-family kinase Fyn has been linked circumstantially to the pathology of Alzheimer's disease, and shown to phosphorylate Tyr18. Recently another Src-family kinase, Lck, has been identified as a genetic risk factor for this disease. Results In this study we show that Lck is a tau kinase. In vitro, comparison of Lck and Fyn showed that while both kinases phosphorylated Tyr18 preferentially, Lck phosphorylated other tyrosines somewhat better than Fyn. In co-transfected COS-7 cells, mutating any one of the five tyrosines in tau to phenylalanine reduced the apparent level of tau tyrosine phosphorylation to 25-40% of that given by wild-type tau. Consistent with this, tau mutants with only one remaining tyrosine gave poor phosphorylation; however, Tyr18 was phosphorylated better than the others. Conclusions Fyn and Lck have subtle differences in their properties as tau kinases, and the phosphorylation of tau is one mechanism by which the genetic risk associated with Lck might be expressed pathogenically.
Collapse
Affiliation(s)
- Timothy Me Scales
- MRC Centre for Neurodegeneration Research, Department of Neuroscience, Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
|
84
|
Lai AY, McLaurin J. Mechanisms of amyloid-Beta Peptide uptake by neurons: the role of lipid rafts and lipid raft-associated proteins. Int J Alzheimers Dis 2010; 2011:548380. [PMID: 21197446 PMCID: PMC3010653 DOI: 10.4061/2011/548380] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 11/29/2010] [Indexed: 12/19/2022] Open
Abstract
A hallmark pathological feature of Alzheimer's disease (AD) is the accumulation of extracellular plaques composed of the amyloid-beta (Aβ) peptide. Thus, classically experiments were designed to examine Aβ toxicities within the central nervous system (CNS) from the extracellular space. However, a significant amount of evidence now suggests that intraneuronal accumulation of Aβ is neurotoxic and may play an important role in the disease progression of AD. One of the means by which neurons accumulate intracellular Aβ is through uptake of extracellular Aβ peptides, and this process may be a potential link between Aβ generation, synaptic dysfunction, and AD pathology. Recent studies have found that neuronal internalization of Aβ involves lipid rafts and various lipid raft-associated receptor proteins. Uptake mechanisms independent of lipid rafts have also been implicated. The aim of this paper is to summarize these findings and discuss their significance in the pathogenesis of AD.
Collapse
Affiliation(s)
- Aaron Y Lai
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 6 Queen's Park Crescent West, Toronto, ON, Canada M5S 3H2
| | | |
Collapse
|
85
|
Chadwick W, Brenneman R, Martin B, Maudsley S. Complex and multidimensional lipid raft alterations in a murine model of Alzheimer's disease. Int J Alzheimers Dis 2010; 2010:604792. [PMID: 21151659 PMCID: PMC2997345 DOI: 10.4061/2010/604792] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 07/27/2010] [Indexed: 01/18/2023] Open
Abstract
Various animal models of Alzheimer's disease (AD) have been created to assist our appreciation of AD pathophysiology, as well as aid development of novel therapeutic strategies. Despite the discovery of mutated proteins that predict the development of AD, there are likely to be many other proteins also involved in this disorder. Complex physiological processes are mediated by coherent interactions of clusters of functionally related proteins. Synaptic dysfunction is one of the hallmarks of AD. Synaptic proteins are organized into multiprotein complexes in high-density membrane structures, known as lipid rafts. These microdomains enable coherent clustering of synergistic signaling proteins. We have used mass analytical techniques and multiple bioinformatic approaches to better appreciate the intricate interactions of these multifunctional proteins in the 3xTgAD murine model of AD. Our results show that there are significant alterations in numerous receptor/cell signaling proteins in cortical lipid rafts isolated from 3xTgAD mice.
Collapse
Affiliation(s)
- Wayne Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Randall Brenneman
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
- Miller School of Medicine, University of Miami, Miami, FL 33124, USA
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| |
Collapse
|
86
|
Nicholson AM, Methner DNR, Ferreira A. Membrane cholesterol modulates {beta}-amyloid-dependent tau cleavage by inducing changes in the membrane content and localization of N-methyl-D-aspartic acid receptors. J Biol Chem 2010; 286:976-86. [PMID: 21047784 DOI: 10.1074/jbc.m110.154138] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously shown that β-amyloid (Aβ) treatment resulted in an age-dependent calpain activation leading to Tau cleavage into a neurotoxic 17-kDa fragment in a cellular model of Alzheimer disease. This detrimental cellular response was mediated by a developmentally regulated increase in membrane cholesterol levels. In this study, we assessed the molecular mechanisms by which cholesterol modulated Aβ-induced Tau cleavage in cultured hippocampal neurons. Our results indicated that these mechanisms did not involve the regulation of the binding of Aβ aggregates to the plasma membrane. On the other hand, experiments using N-methyl-d-aspartic acid receptor inhibitors suggested that these receptors played an essential role in cholesterol-mediated Aβ-dependent calpain activity and 17-kDa Tau production. Biochemical and immunocytochemical analyses demonstrated that decreasing membrane cholesterol levels in mature neurons resulted in a significant reduction of the NR1 subunit at the membrane as well as an increase in the number of large NR1, NR2A, and NR2B subunit clusters. Moreover, the majority of these larger N-methyl-d-aspartic acid receptor subunit immunoreactive spots was not juxtaposed to presynaptic sites in cholesterol-reduced neurons. These data suggested that changes at the synaptic level underlie the mechanism by which membrane cholesterol modulates developmental changes in the susceptibility of hippocampal neurons to Aβ-induced toxicity.
Collapse
Affiliation(s)
- Alexandra M Nicholson
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
87
|
Watts HR, Anderson PJB, Ma D, Philpott KL, Jen SM, Croucher M, Jen LS, Gentleman SM. Differential effects of amyloid-β peptide aggregation status on in vivo retinal neurotoxicity. Eye Brain 2010; 2:121-137. [PMID: 28539771 PMCID: PMC5436173 DOI: 10.2147/eb.s9902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The present study examined the relationship between amyloid beta (Aβ)-peptide aggregation state and neurotoxicity in vivo using the rat retinal-vitreal model. Following single unilateral intravitreal injection of either soluble Aβ1-42 or Aβ1-42 preaggregated for different periods, retinal pathology was evaluated at 24 hours, 48 hours, and 1-month postinjection. Injection of either soluble Aβ (sAβ) or preaggregated Aβ induced a rapid reduction in immunoreactivity (IR) for synaptophysin, suggesting that direct contact with neurons is not necessary to disrupt synapses. Acute neuronal ionic and metabolic dysfunction was demonstrated by widespread loss of IR to the calcium buffering protein parvalbumin (PV) and protein gene product 9.5, a component of the ubiquitin-proteosome system. Injection of sAβ appeared to have a more rapid impact on PV than the preaggregated treatments, producing a marked reduction in PV cell diameters at 48 hours, an effect that was only observed for preaggregated Aβ after 1-month survival. Extending the preaggregation period from 4 to 8 days to obtain highly fibrillar Aβ species significantly increased the loss of choline acteyltransferase IR, but had no effect on PV-IR. These findings prompt the conclusion that Aβ assembly state has a significant impact on in vivo neurotoxicity by triggering distinct molecular changes within the cell.
Collapse
Affiliation(s)
- HR Watts
- Department of Medicine, Imperial College London, Charing Cross Campus, London, UK
| | - PJB Anderson
- Department of Medicine, Imperial College London, Charing Cross Campus, London, UK
| | - D Ma
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - KL Philpott
- Neurosciences, Centre of Excellence for Drug Discovery, GlaxoSmithKline Pharmaceuticals, Harlow, Essex, UK
| | - SM Jen
- Department of Medicine, Imperial College London, Charing Cross Campus, London, UK
| | - M Croucher
- Department of Medicine, Imperial College London, Charing Cross Campus, London, UK
| | - LS Jen
- Department of Medicine, Imperial College London, Charing Cross Campus, London, UK
| | - SM Gentleman
- Department of Medicine, Imperial College London, Charing Cross Campus, London, UK
| |
Collapse
|
88
|
Haughey NJ, Bandaru VVR, Bae M, Mattson MP. Roles for dysfunctional sphingolipid metabolism in Alzheimer's disease neuropathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1801:878-86. [PMID: 20452460 PMCID: PMC2907186 DOI: 10.1016/j.bbalip.2010.05.003] [Citation(s) in RCA: 211] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 04/29/2010] [Accepted: 05/03/2010] [Indexed: 12/12/2022]
Abstract
Sphingolipids in the membranes of neurons play important roles in signal transduction, either by modulating the localization and activation of membrane-associated receptors or by acting as precursors of bioactive lipid mediators. Activation of cytokine and neurotrophic factor receptors coupled to sphingomyelinases results in the generation of ceramides and gangliosides, which in turn, modify the structural and functional plasticity of neurons. In aging and neurodegenerative conditions such as Alzheimer's disease (AD), there are increased membrane-associated oxidative stress and excessive production and accumulation of ceramides. Studies of brain tissue samples from human subjects, and of experimental models of the diseases, suggest that perturbed sphingomyelin metabolism is a pivotal event in the dysfunction and degeneration of neurons that occurs in AD and HIV dementia. Dietary and pharmacological interventions that target sphingolipid metabolism should be pursued for the prevention and treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | |
Collapse
|
89
|
Pooler AM, Hanger DP. Functional implications of the association of tau with the plasma membrane. Biochem Soc Trans 2010; 38:1012-5. [PMID: 20658995 DOI: 10.1042/bst0381012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Tau is an abundant microtubule-associated protein which regulates the stability of the cytoskeleton. Tau binds microtubules directly through microtubule-binding domains in its C-terminus. However, tau is not only located in the cytosol of cells, but also associated with other intracellular domains, including the plasma membrane, suggesting that tau may have additional functions other than stabilizing the neuronal cytoskeleton. Localization of tau at the cell surface appears to be dependent on interactions of the N-terminal projection domain of tau. Furthermore, membrane-associated tau is dephosphorylated at serine/threonine residues, suggesting that the phosphorylation state of tau regulates its intracellular trafficking. Dephosphorylation of tau may increase the association of tau with trafficking proteins which target tau to the plasma membrane. Thus it is possible that the hyperphosphoryation of tau may contribute to the pathogenesis of Alzheimer's disease by promoting the formation of neurofibrillary tangles from cytosolic tau, and also by inhibiting additional tau functions through disruption of its targeting to the plasma membrane.
Collapse
Affiliation(s)
- Amy M Pooler
- Department of Neuroscience, Institute of Psychiatry, King's College London, MRC Centre for Neurodegeneration Research, De Crespigny Park, London SE5 8AF, UK.
| | | |
Collapse
|
90
|
Nishimura AL, Zupunski V, Troakes C, Kathe C, Fratta P, Howell M, Gallo JM, Hortobágyi T, Shaw CE, Rogelj B. Nuclear import impairment causes cytoplasmic trans-activation response DNA-binding protein accumulation and is associated with frontotemporal lobar degeneration. Brain 2010; 133:1763-71. [PMID: 20472655 DOI: 10.1093/brain/awq111] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Trans-activation response DNA-binding protein (TDP-43) accumulation is the major component of ubiquitinated protein inclusions found in patients with amyotrophic lateral sclerosis, and frontotemporal lobar degeneration with TDP-43 positive ubiquitinated inclusions, recently relabelled the 'TDP-43 proteinopathies'. TDP-43 is predominantly located in the nucleus, however, in disease it mislocalizes to the cytoplasm where it aggregates to form hallmark pathological inclusions. The identification of TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis cases confirms its pathogenic role; but it is wild-type TDP-43 that is deposited in the vast majority of TDP-43 proteinopathies, implicating other unknown factors for its mislocalization and aggregation. One such mechanism may be defective nuclear import of TDP-43 protein, as a disruption of its nuclear localization signal leads to mislocalization and aggregation of TDP-43 in the cytoplasm. In order to explore the factors that regulate the nuclear import of TDP-43, we used a small interfering RNA library to silence 82 proteins involved in nuclear transport and found that knockdowns of karyopherin-beta1 and cellular apoptosis susceptibility protein resulted in marked cytoplasmic accumulation of TDP-43. In glutathione S-transferase pull-down assays, TDP-43 bound to karyopherin-alphas, thereby confirming the classical nuclear import pathway for the import of TDP-43. Analysis of the expression of chosen nuclear import factors in post-mortem brain samples from patients with TDP-43 positive frontotemporal lobar degeneration, and spinal cord samples from patients with amyotrophic lateral sclerosis, revealed a considerable reduction in expression of cellular apoptosis susceptibility protein in frontotemporal lobar degeneration. We propose that cellular apoptosis susceptibility protein associated defective nuclear transport may play a mechanistic role in the pathogenesis of the TDP-43 positive frontotemporal lobar degeneration.
Collapse
Affiliation(s)
- Agnes L Nishimura
- Medical Research Council (MRC) Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, London, SE5 8AF, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Membrane biophysics and mechanics in Alzheimer's disease. Mol Neurobiol 2010; 41:138-48. [PMID: 20437210 DOI: 10.1007/s12035-010-8121-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 03/17/2010] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease is a chronic neurodegenerative disorder characterized by neuronal loss, cerebrovascular inflammation, and accumulation of senile plaques in the brain parenchyma and cerebral blood vessels. Amyloid-beta peptide (Abeta), a major component of senile plaques, has been shown to exert multiple toxic effects to neurons, astrocytes, glial cells, and brain endothelium. Oligomeric Abeta can disturb the structure and function of cell membranes and alter membrane mechanical properties, such as membrane fluidity and molecular order. Much of these effects are attributed to their capability to trigger oxidative stress and inflammation. In this review, we discuss the effects of Abeta on neuronal cells, astrocytes, and cerebral endothelial cells with special emphasis on cell membrane properties and cell functions.
Collapse
|
92
|
Schengrund CL. Lipid rafts: Keys to neurodegeneration. Brain Res Bull 2010; 82:7-17. [DOI: 10.1016/j.brainresbull.2010.02.013] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 02/16/2010] [Accepted: 02/23/2010] [Indexed: 01/11/2023]
|
93
|
Zampagni M, Evangelisti E, Cascella R, Liguri G, Becatti M, Pensalfini A, Uberti D, Cenini G, Memo M, Bagnoli S, Nacmias B, Sorbi S, Cecchi C. Lipid rafts are primary mediators of amyloid oxidative attack on plasma membrane. J Mol Med (Berl) 2010; 88:597-608. [DOI: 10.1007/s00109-010-0603-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 02/10/2010] [Accepted: 02/17/2010] [Indexed: 12/14/2022]
|
94
|
Hanger DP, Seereeram A, Noble W. Mediators of tau phosphorylation in the pathogenesis of Alzheimer's disease. Expert Rev Neurother 2010; 9:1647-66. [PMID: 19903024 DOI: 10.1586/ern.09.104] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The need for disease-modifying drugs for Alzheimer's disease has become increasingly important owing to escalating disease prevalence and the associated socio-economic burden. Until recently, reducing brain amyloid accumulation has been the main therapeutic focus; however, increasing evidence suggests that targeting abnormal tau phosphorylation could be beneficial. Tau is phosphorylated by several protein kinases and this is balanced by dephosphorylation by protein phosphatases. Phosphorylation at specific sites can influence the physiological functions of tau, including its role in binding to and stabilizing the neuronal cytoskeleton. aberrant phosphorylation of tau could render it susceptible to potentially pathogenic alterations, including conformational changes, proteolytic cleavage and aggregation. While strategies that reduce tau phosphorylation in transgenic models of disease have been promising, our understanding of the mechanisms through which tau becomes abnormally phosphorylated in disease is lacking.
Collapse
Affiliation(s)
- Diane P Hanger
- MRC Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, Department of Neuroscience (P037), De Crespigny Park, London SE5 8AF, UK.
| | | | | |
Collapse
|
95
|
Cecchi C, Nichino D, Zampagni M, Bernacchioni C, Evangelisti E, Pensalfini A, Liguri G, Gliozzi A, Stefani M, Relini A. A protective role for lipid raft cholesterol against amyloid-induced membrane damage in human neuroblastoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2204-16. [DOI: 10.1016/j.bbamem.2009.07.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 07/16/2009] [Accepted: 07/23/2009] [Indexed: 12/14/2022]
|
96
|
Crews L, Tsigelny I, Hashimoto M, Masliah E. Role of synucleins in Alzheimer's disease. Neurotox Res 2009; 16:306-17. [PMID: 19551456 PMCID: PMC2727399 DOI: 10.1007/s12640-009-9073-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 05/07/2009] [Accepted: 06/08/2009] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common causes of dementia and movement disorders in the elderly. While progressive accumulation of oligomeric amyloid-beta protein (Abeta) has been identified as one of the central toxic events in AD leading to synaptic dysfunction, accumulation of alpha-synuclein (alpha-syn) resulting in the formation of oligomers has been linked to PD. Most of the studies in AD have been focused on investigating the role of Abeta and Tau; however, recent studies suggest that alpha-syn might also play a role in the pathogenesis of AD. For example, fragments of alpha-syn can associate with amyloid plaques and Abeta promotes the aggregation of alpha-syn in vivo and worsens the deficits in alpha-syn tg mice. Moreover, alpha-syn has also been shown to accumulate in limbic regions in AD, Down's syndrome, and familial AD cases. Abeta and alpha-syn might directly interact under pathological conditions leading to the formation of toxic oligomers and nanopores that increase intracellular calcium. The interactions between Abeta and alpha-syn might also result in oxidative stress, lysosomal leakage, and mitochondrial dysfunction. Thus, better understanding the steps involved in the process of Abeta and alpha-syn aggregation is important in order to develop intervention strategies that might prevent or reverse the accumulation of toxic proteins in AD.
Collapse
Affiliation(s)
- Leslie Crews
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093-0624 USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92093-0624 USA
| | - Igor Tsigelny
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0624 USA
- San Diego Super Computer Center, University of California San Diego, La Jolla, CA 92093-0624 USA
| | - Makoto Hashimoto
- Laboratory for Chemistry and Metabolism, Tokyo Metropolitan Institute for Neuroscience, Fuchu, Tokyo, Japan
| | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093-0624 USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92093-0624 USA
| |
Collapse
|
97
|
Gaspar RC, Villarreal SA, Bowles N, Hepler RW, Joyce JG, Shughrue PJ. Oligomers of beta-amyloid are sequestered into and seed new plaques in the brains of an AD mouse model. Exp Neurol 2009; 223:394-400. [PMID: 19744481 DOI: 10.1016/j.expneurol.2009.09.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 08/19/2009] [Accepted: 09/01/2009] [Indexed: 11/27/2022]
Abstract
Amyloid plaque deposition in the brain is a hallmark of Alzheimer's disease, but recent evidence indicates that the disease may be primarily caused by soluble amyloid-beta (1-42) (Abeta) oligomers or Abeta-derived diffusible ligands (ADDLs). ADDLs induce cognitive deficits in animal models and are thought to assemble in vitro by a mechanism apart from plaque formation. To investigate the in vivo relationship of ADDLs and plaques, biotin-labeled ADDLs (bADDLs) or amylin oligomers (bAMs) were injected into the hippocampus of hAPP overexpressing mice. The brains were collected 1 or 5 weeks after the last treatment and were processed for immunohistochemistry. Staining of tissue 1 week post-treatment showed bADDLs had diffused throughout the tissue and incorporated into plaques. Additionally, small deposits of thioflavin S-negative bADDLs were observed. At 5 weeks post-treatment, thioflavin S-positive material continued to accumulate around plaques containing bADDLs. Thioflavin S-positive material also accrued around bADDL deposits, implying that bADDLs were capable of seeding new plaques. In contrast, bAMs cleared from the brain and did not accumulate in plaques. Together, these data indicate that ADDLs are able to contribute to in vivo plaque formation in a peptide-specific manner.
Collapse
Affiliation(s)
- Renee C Gaspar
- Department of Integrative Systems Neuroscience, West Point, PA, USA.
| | | | | | | | | | | |
Collapse
|
98
|
Thompson AJ, Williamson R, Schofield E, Stephenson J, Hanger D, Anderton B. Quantitation of glycogen synthase kinase-3 sensitive proteins in neuronal membrane rafts. Proteomics 2009; 9:3022-35. [PMID: 19526546 DOI: 10.1002/pmic.200900006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 02/18/2009] [Indexed: 12/26/2022]
Abstract
We report a quantitative proteomic study to investigate the changes induced in membrane rafts by the inhibition of glycogen synthase kinase-3. Sensitive quantitation of membrane raft proteins using isobaric tagging chemistries was enabled by a novel hybrid proteomic method to isolate low-microgram (10-30 microg) membrane raft protein preparations as unresolved bands in a low-density acrylamide gel. Samples were in-gel digested, differentially tagged and combined for 2-D LC and quantitative MS. Analysis of hippocampal membrane preparations using this approach resulted in a sixfold increase in sensitivity and a threefold increase in the number of quantifiable proteins compared with parallel processing using a traditional in-solution method. Quantitative analysis of membrane raft preparations from a human neuronal cell line treated with glycogen synthase kinase-3 inhibitors SB415286 or lithium chloride, that have been reported to modulate processing of the Alzheimer amyloid precursor protein, identified several protein changes. These included decreases in lamin B1 and lamin B receptor, as well as increases in several endosome regulating rab proteins, rab5, rab7 and rab11 that have been implicated in processing of the amyloid precursor protein in Alzheimer's disease.
Collapse
Affiliation(s)
- Andrew J Thompson
- Department of Neuroscience, MRC Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, London, UK.
| | | | | | | | | | | |
Collapse
|
99
|
Hanger DP, Anderton BH, Noble W. Tau phosphorylation: the therapeutic challenge for neurodegenerative disease. Trends Mol Med 2009; 15:112-9. [PMID: 19246243 DOI: 10.1016/j.molmed.2009.01.003] [Citation(s) in RCA: 704] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 01/09/2009] [Accepted: 01/09/2009] [Indexed: 01/22/2023]
Abstract
The microtubule-associated protein tau is integral to the pathogenesis of Alzheimer's disease (AD), as well as several related disorders, termed tauopathies, in which tau is deposited in affected brain regions. In the tauopathies, pathological tau is in an elevated state of phosphorylation and is aberrantly cleaved. It also exhibits abnormal conformations and becomes aggregated, resulting in neurofibrillary tau pathology. Recent evidence suggests that relatively early disease-associated changes in soluble tau proteins, including phosphorylation, are involved in the induction of neuronal death. Here, we summarize recent developments that suggest new therapeutic strategies to prevent or reduce the progression of pathology in the tauopathies. A list of tau phosphorylation sites identified in the tauopathies and in controls accompanies this review.
Collapse
Affiliation(s)
- Diane P Hanger
- MRC Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, UK.
| | | | | |
Collapse
|
100
|
Sondag CM, Dhawan G, Combs CK. Beta amyloid oligomers and fibrils stimulate differential activation of primary microglia. J Neuroinflammation 2009; 6:1. [PMID: 19123954 PMCID: PMC2632990 DOI: 10.1186/1742-2094-6-1] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 01/05/2009] [Indexed: 12/31/2022] Open
Abstract
Background Beta amyloid (Aβ) peptides are the major constituents of the senile plaques present in Alzheimer's diseased brain. Pathogenesis has been associated with the aggregated form of the peptide as these fibrils are the conformation readily found in the plaques. However, recent studies have shown that the nonaggregated, soluble assemblies of Aβ have the potential to stimulate neuronal dysfunction and may play a prominent role in the pathogenesis of Alzheimer's disease. Methods Soluble, synthetic Aβ1–42 oligomers were prepared producing mainly dimer-trimer conformations as assessed by SDS-PAGE. Similar analysis demonstrated fibril preparations to produce large insoluble aggregates unable to migrate out of the stacking portion of the gels. These peptide preparations were used to stimulate primary murine microglia and cortical neuron cultures. Microglia were analyzed for changes in signaling response and secretory phenotype via Western analysis and ELISA. Viability was examined by quantifying lactate dehydrogenase release from the cultures. Results Aβ oligomers and fibrils were used to stimulate microglia for comparison. Both the oligomers and fibrils stimulated proinflammatory activation of primary microglia but the specific conformation of the peptide determined the activation profile. Oligomers stimulated increased levels of active, phosphorylated Lyn and Syk kinase as well as p38 MAP kinase compared to fibrils. Moreover, oligomers stimulated a differential secretory profile for interleukin 6, monocyte chemoattractant protein-1 and keratinocyte chemoattractant when compared to fibrils. Finally, soluble oligomers stimulated death of cultured cortical neurons that was exacerbated by the presence of microglia. Conclusion These data suggest that fibrils and oligomers stimulate unique signaling responses in microglia leading to discrete secretory changes and effects on neuron survival. This suggests that inflammation changes during disease may be the consequence of unique peptide-stimulated events and each conformation may represent an individual anti-inflammatory therapeutic target.
Collapse
Affiliation(s)
- Cindy M Sondag
- Department of Pharmacology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA.
| | | | | |
Collapse
|