101
|
Boddapati S, Levites Y, Sierks MR. Inhibiting β-secretase activity in Alzheimer's disease cell models with single-chain antibodies specifically targeting APP. J Mol Biol 2010; 405:436-47. [PMID: 21073877 DOI: 10.1016/j.jmb.2010.10.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/27/2010] [Accepted: 10/27/2010] [Indexed: 01/09/2023]
Abstract
The Amyloid-β (Aβ) peptide is produced from the amyloid precursor protein (APP) by sequential proteolytic cleavage of APP first by β-secretase and then by γ-secretase. β-Site APP cleaving enzyme-1 (BACE-1) is the predominant enzyme involved in β-secretase processing of APP and is a primary therapeutic target for treatment of Alzheimer's disease. While inhibiting BACE-1 activity has obvious therapeutic advantages, BACE-1 also cleaves numerous other substrates with important physiological activity. Thus, blanket inhibition of BACE-1 function may have adverse side effects. We isolated a single chain variable fragment (scFv) from a human-based scFv yeast display library that selectively inhibits BACE-1 activity toward APP by binding the APP substrate at the proteolytic site. We selected the iBSEC1 scFv, since it recognizes the BACE-1 cleavage site on APP but does not bind the adjacent highly antigenic N-terminal of Aβ, and thus it will target APP but not soluble Aβ. When added to 7PA2 cells, a mammalian cell line that overexpresses APP, the iBSEC1 scFv binds APP on the cell surface, reduces toxicity induced by APP overexpression, and reduces both intracellular and extracellular Aβ levels by around 50%. Since the iBSEC1 scFv does not contain the antibody F(c) region, this construct does not pose the risk of exacerbating inflammation in the brain as faced with full-length monoclonal antibodies for potential therapeutic applications.
Collapse
Affiliation(s)
- Shanta Boddapati
- Department of Chemical Engineering, Arizona State University, Tempe, AZ 85287-6106, USA
| | | | | |
Collapse
|
102
|
Vingtdeux V, Chandakkar P, Zhao H, d'Abramo C, Davies P, Marambaud P. Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-β peptide degradation. FASEB J 2010; 25:219-31. [PMID: 20852062 DOI: 10.1096/fj.10-167361] [Citation(s) in RCA: 182] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AMP-activated protein kinase (AMPK) is a metabolic sensor involved in intracellular energy metabolism through the control of several homeostatic mechanisms, which include autophagy and protein degradation. Recently, we reported that AMPK activation by resveratrol promotes autophagy-dependent degradation of the amyloid-β (Aβ) peptides, the core components of the cerebral senile plaques in Alzheimer's disease. To identify more potent enhancers of Aβ degradation, we screened a library of synthetic small molecules selected for their structural similarities with resveratrol. Here, we report the identification of a series of structurally related molecules, the RSVA series, which inhibited Aβ accumulation in cell lines nearly 40 times more potently than did resveratrol. Two of these molecules, RSVA314 and RSVA405, were further characterized and were found to facilitate CaMKKβ-dependent activation of AMPK, to inhibit mTOR (mammalian target of rapamycin), and to promote autophagy to increase Aβ degradation by the lysosomal system (apparent EC(50) ∼ 1 μM). This work identifies the RSVA compounds as promising lead molecules for the development of a new class of AMPK activating drugs controlling mTOR signaling, autophagy, and Aβ clearance.
Collapse
Affiliation(s)
- Valérie Vingtdeux
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institute for Medical Research, North Shore-Long Island Jewish Medical Center, Manhasset, New York, NY 11030, USA
| | | | | | | | | | | |
Collapse
|
103
|
Singh TD, Park SY, Bae JS, Yun Y, Bae YC, Park RW, Kim IS. MEGF10 functions as a receptor for the uptake of amyloid-β. FEBS Lett 2010; 584:3936-42. [DOI: 10.1016/j.febslet.2010.08.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 07/31/2010] [Accepted: 08/30/2010] [Indexed: 10/19/2022]
|
104
|
Nie HZ, Shi S, Lukas RJ, Zhao WJ, Sun YN, Yin M. Activation of α7 nicotinic receptor affects APP processing by regulating secretase activity in SH-EP1-α7 nAChR-hAPP695 cells. Brain Res 2010; 1356:112-20. [PMID: 20708605 DOI: 10.1016/j.brainres.2010.07.110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 07/30/2010] [Accepted: 07/31/2010] [Indexed: 12/31/2022]
Abstract
Multiple lines of evidence have implicated that nicotinic acetylcholine receptor (nAChR) may be an important therapeutic target for the treatment of Alzheimer's disease (AD). Although there are reports suggesting a link between alpha7 nAChR subtype and AD, there has been little report on the mechanism. The present study investigates whether and how α7 nAChR activation affects APP695 processing in SH-EP1 cell model. Cell line co-expressing α7 nAChR gene and human amyloid precursor protein 695 (hAPP695) gene were constructed by stable transfection. Expression of β-amyloid, α-form of secreted APP (αAPPs) and APP1695 was measured by ELISA, western blotting and real-time PCR respectively. Additionally, α, β, and γ-secretase activities were also analyzed in constructed SH-EP1-α7 nAChR-hAPP695 cell line. The results showed that SH-EP1-α7 nAChR-hAPP695 cell line, expressing both hAPP695 gene and α7 nAChR subtype gene, was constructed successfully. The secreted Aβ was decreased and αAPPs was significantly increased by non-selective nAChR agonist nicotine (10 μM) and specific α7 nAChR agonist GTS-21 (1 μM), and APP expression was not affected. Furthermore, specific α7 nAChR antagonist methyllycaconitine (MLA) reversed the alterations induced by activation of α7 nAChR. CTF-α was increased and CTF-γ was decreased when treated with nicotine (10 μM). In addition, the results of enymatic activity analysis showed that nicotine (1μM) and GTS-21 (0.1, 1 μM) decreased γ-secretase activity, but has no effects on α-secretase activity and β-secretase activity. Our findings demonstrate that, through regulating γ-secretase activity, α7 nAChR activation reduces APP processing in amyloidogenic pathway, and at the same time enhances APP processing in non-amyloidogenic pathway. The constructed SH-EP1-α7 nAChR-hAPP695 cell line might be useful for screening specific nAChR agonists against AD.
Collapse
Affiliation(s)
- Hui Zhen Nie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | | | | | | | | | | |
Collapse
|
105
|
Hanessian S, Maji DK, Govindan S, Matera R, Tintelnot-Blomley M. Substrate-controlled and organocatalytic asymmetric synthesis of carbocyclic amino acid dipeptide mimetics. J Org Chem 2010; 75:2861-76. [PMID: 20392053 DOI: 10.1021/jo100017t] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The asymmetric synthesis of a carbocyclic delta-amino acid representing the P(2)/P(3) subunit of a nonpeptidic truncated peptidomimetic molecule is described relying on two independent approaches.
Collapse
Affiliation(s)
- Stephen Hanessian
- Department of Chemistry, Université de Montréal, P.O. Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada.
| | | | | | | | | |
Collapse
|
106
|
Loss of Hsp110 leads to age-dependent tau hyperphosphorylation and early accumulation of insoluble amyloid beta. Mol Cell Biol 2010; 30:4626-43. [PMID: 20679486 DOI: 10.1128/mcb.01493-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Accumulation of tau into neurofibrillary tangles is a pathological consequence of Alzheimer's disease and other tauopathies. Failures of the quality control mechanisms by the heat shock proteins (Hsps) positively correlate with the appearance of such neurodegenerative diseases. However, in vivo genetic evidence for the roles of Hsps in neurodegeneration remains elusive. Hsp110 is a nucleotide exchange factor for Hsp70, and direct substrate binding to Hsp110 may facilitate substrate folding. Hsp70 complexes have been implicated in tau phosphorylation state and amyloid precursor protein (APP) processing. To provide evidence for a role for Hsp110 in central nervous system homeostasis, we have generated hsp110(-)(/)(-) mice. Our results show that hsp110(-)(/)(-) mice exhibit accumulation of hyperphosphorylated-tau (p-tau) and neurodegeneration. We also demonstrate that Hsp110 is in complexes with tau, other molecular chaperones, and protein phosphatase 2A (PP2A). Surprisingly, high levels of PP2A remain bound to tau but with significantly reduced activity in brain extracts from aged hsp110(-)(/)(-) mice compared to brain extracts from wild-type mice. Mice deficient in the Hsp110 partner (Hsp70) also exhibit a phenotype comparable to that of hsp110(-)(/)(-) mice, confirming a critical role for Hsp110-Hsp70 in maintaining tau in its unphosphorylated form during aging. In addition, crossing hsp110(-)(/)(-) mice with mice overexpressing mutant APP (APPβsw) leads to selective appearance of insoluble amyloid β42 (Aβ42), suggesting an essential role for Hsp110 in APP processing and Aβ generation. Thus, our findings provide in vivo evidence that Hsp110 plays a critical function in tau phosphorylation state through maintenance of efficient PP2A activity, confirming its role in pathogenesis of Alzheimer's disease and other tauopathies.
Collapse
|
107
|
Ramesh BN, Rao TSS, Prakasam A, Sambamurti K, Rao KSJ. Neuronutrition and Alzheimer's disease. J Alzheimers Dis 2010; 19:1123-39. [PMID: 20308778 DOI: 10.3233/jad-2010-1312] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder resulting from both genetic and environmental factors with the latter being particularly important for the sporadic form of the disease. As such, diets rich in saturated fatty acids and alcohol, and deficient in antioxidants and vitamins appear to promote the onset of the disease, while diets rich in unsaturated fatty acids, vitamins, antioxidants, and wine likely suppress its onset. In addition, evidence suggests that diets rich in polyphenols and some spices suppress the onset of AD by scavenging free radicals and preventing oxidative damage. Metal ions are known to catalyze the production of free radicals and induce mental retardation or dementia, and several studies have also identified metals such as Pb, Fe, Al, Cu, and Zn in AD pathogenesis. While specific metal chelators have been tested for therapy, they have not been very successful, probably due to their late administration, i.e., after brain damage has been triggered. Since several dietary polyphenols are known to chelate metals, their routine use may also be protective against the onset of AD. In this review, we summarize beneficial dietary techniques in the fight against AD.
Collapse
Affiliation(s)
- Balenahalli N Ramesh
- Biochemistry and Nutrition, Central Food Technological Research Institute, CSIR Unit, Mysore, India
| | | | | | | | | |
Collapse
|
108
|
Zhang L, Yu H, Zhao X, Lin X, Tan C, Cao G, Wang Z. Neuroprotective effects of salidroside against beta-amyloid-induced oxidative stress in SH-SY5Y human neuroblastoma cells. Neurochem Int 2010; 57:547-55. [PMID: 20615444 DOI: 10.1016/j.neuint.2010.06.021] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 06/24/2010] [Indexed: 01/26/2023]
Abstract
Beta-amyloid (Abeta) peptide, the hallmark of Alzheimer's disease (AD), invokes a cascade of oxidative damages to neurons and eventually leads to neuronal death. In this study, salidroside (Sald), an active compound isolated from a traditional Chinese medicinal plant, Rhodiola rosea L., was investigated to assess its protective effects and the underlying mechanisms against Abeta-induced oxidative stress in SH-SY5Y human neuroblastoma cells. Abeta(25-35)-induced neuronal toxicity was characterized by the decrease of cell viability, the release of lactate dehydrogenase (LDH), morphological alterations, neuronal DNA condensation, and the cleavage of poly(ADP-ribose) polymerase (PARP) by activated caspase-3. Pretreatment with salidroside markedly attenuated Abeta(25-35)-induced loss of cell viability and apoptosis in a dose-dependent manner. The mechanisms of salidroside protected neurons from oxidative stress included the induction of antioxidant enzymes, thioredoxin (Trx), heme oxygenase-1 (HO-1), and peroxiredoxin-I (PrxI); the downregulation of pro-apoptotic protein Bax and the upregulation of anti-apoptotic protein Bcl-X(L). Furthermore, salidroside dose-dependently restored Abeta(25-35)-induced loss of mitochondrial membrane potential (MMP) as well as suppressed the elevation of intracellular reactive oxygen species (ROS) level. It was also observed that Abeta(25-35) stimulated the phosphorylation of mitogen-activated protein (MAP) kinases, including c-Jun NH(2)-terminal kinase (JNK) and p38 MAP kinase, but not extracellular signal-regulated kinase1/2 (ERK1/2). Salidroside inhibited Abeta(25-35)-induced phosphorylation of JNK and p38 MAP kinase, but not ERK1/2. These results suggest that salidroside has protective effects against Abeta(25-35)-induced oxidative stress, which might be a potential therapeutic agent for treating or preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Zhang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
109
|
Bergmans BA, Shariati SAM, Habets RLP, Verstreken P, Schoonjans L, Müller U, Dotti CG, De Strooper B. Neurons generated from APP/APLP1/APLP2 triple knockout embryonic stem cells behave normally in vitro and in vivo: lack of evidence for a cell autonomous role of the amyloid precursor protein in neuronal differentiation. Stem Cells 2010; 28:399-406. [PMID: 20049903 DOI: 10.1002/stem.296] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Alzheimer's disease amyloid precursor protein (APP) has been implicated in many neurobiologic processes, but supporting evidence remains indirect. Studies are confounded by the existence of two partially redundant APP homologues, APLP1 and APLP2. APP/APLP1/APLP2 triple knockout (APP tKO) mice display cobblestone lissencephaly and are perinatally lethal. To circumvent this problem, we generated APP triple knockout embryonic stem (ES) cells and differentiated these to APP triple knockout neurons in vitro and in vivo. In comparison with wild-type (WT) ES cell-derived neurons, APP tKO neurons formed equally pure neuronal cultures, had unaltered in vitro migratory capacities, had a similar acquisition of polarity, and were capable of extending long neurites and forming active excitatory synapses. These data were confirmed in vivo in chimeric mice with APP tKO neurons expressing the enhanced green fluorescent protein (eGFP) present in a WT background brain. The results suggest that the loss of the APP family of proteins has no major effect on these critical neuronal processes and that the apparent multitude of functions in which APP has been implicated might be characterized by molecular redundancy. Our stem cell culture provides an excellent tool to circumvent the problem of lack of viability of APP/APLP triple knockout mice and will help to explore the function of this intriguing protein further in vitro and in vivo.
Collapse
Affiliation(s)
- Bruno A Bergmans
- Laboratory of Neuronal Cell Biology and Gene Transfer, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Niikura T, Tajima H, Kita Y. Neuronal cell death in Alzheimer's disease and a neuroprotective factor, humanin. Curr Neuropharmacol 2010; 4:139-47. [PMID: 18615127 DOI: 10.2174/157015906776359577] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Revised: 11/09/2005] [Accepted: 12/08/2005] [Indexed: 11/22/2022] Open
Abstract
Brain atrophy caused by neuronal loss is a prominent pathological feature of Alzheimer's disease (AD). Amyloid beta (Abeta), the major component of senile plaques, is considered to play a central role in neuronal cell death. In addition to removal of the toxic Abeta, direct suppression of neuronal loss is an essential part of AD treatment; however, no such neuroprotective therapies have been developed. Excess amount of Abeta evokes multiple cytotoxic mechanisms, involving increase of the intracellular Ca(2+) level, oxidative stress, and receptor-mediated activation of cell-death cascades. Such diversity in cytotoxic mechanisms induced by Abeta clearly indicates a complex nature of the AD-related neuronal cell death. We have identified a 24-residue peptide, Humanin (HN), which suppresses in vitro neuronal cell death caused by all AD-related insults, including Abeta, so far tested. The anti-AD effect of HN has been further confirmed in vivo using mice with Abeta-induced amnesia. Altogether, such potent neuroprotective activity of HN against AD-relevant cytotoxicity both in vitro and in vivo suggests the potential clinical applications of HN in novel AD therapies aimed at controlling neuronal death.
Collapse
Affiliation(s)
- Takako Niikura
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Tokyo 160-8582, Japan.
| | | | | |
Collapse
|
111
|
Effects of nonsteroidal anti-inflammatory drugs on amyloid-beta pathology in mouse skeletal muscle. Neurobiol Dis 2010; 39:449-56. [PMID: 20493261 DOI: 10.1016/j.nbd.2010.05.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Revised: 04/26/2010] [Accepted: 05/11/2010] [Indexed: 10/19/2022] Open
Abstract
Sporadic inclusion body myositis (sIBM) is a common age-related inflammatory myopathy characterized by the presence of intracellular inclusions that contain the amyloid-beta (Abeta) peptide, a derivative of the amyloid precursor protein (APP). Abeta is believed to cause Alzheimer's disease (AD), suggesting that a link may exist between the two diseases. If AD and sIBM are linked, then treatments that lower Abeta in brain may prove useful for sIBM. To test this hypothesis, transgenic mice that overexpress APP in skeletal muscle were treated for 6 months with a variety of nonsteroidal anti-inflammatory drugs (NSAIDs; naproxen, ibuprofen, carprofen or R-flurbiprofen), a subset of which reduce Abeta in brain and cultured cells. Only ibuprofen lowered Abeta in muscle, and this was not accompanied by corresponding improvements in phenotype. These results indicate that the effects of NSAIDs in the brain may be different from other tissues and that Abeta alone cannot account for skeletal muscle dysfunction in these mice.
Collapse
|
112
|
Oliveira TG, Di Paolo G. Phospholipase D in brain function and Alzheimer's disease. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:799-805. [PMID: 20399893 DOI: 10.1016/j.bbalip.2010.04.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 04/07/2010] [Accepted: 04/08/2010] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease is the most common neurodegenerative disorder. Although lipids are major constituents of brain, their role in Alzheimer's disease pathogenesis is poorly understood. Much attention has been given to cholesterol, but growing evidence suggests that other lipids, such as phospholipids, might play an important role in this disorder. In this review, we will summarize the evidence linking phospholipase D, a phosphatidic acid-synthesizing enzyme, to multiple aspects of normal brain function and to Alzheimer's disease. The role of phospholipase D in signaling mechanisms downstream of beta-amyloid as well as in the trafficking and processing of amyloid precursor protein will be emphasized.
Collapse
Affiliation(s)
- Tiago Gil Oliveira
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | | |
Collapse
|
113
|
Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE, Janle EM, Lobo J, Ferruzzi MG, Davies P, Marambaud P. AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J Biol Chem 2010; 285:9100-13. [PMID: 20080969 PMCID: PMC2838330 DOI: 10.1074/jbc.m109.060061] [Citation(s) in RCA: 486] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 12/02/2009] [Indexed: 02/03/2023] Open
Abstract
Alzheimer disease is an age-related neurodegenerative disorder characterized by amyloid-beta (Abeta) peptide deposition into cerebral amyloid plaques. The natural polyphenol resveratrol promotes anti-aging pathways via the activation of several metabolic sensors, including the AMP-activated protein kinase (AMPK). Resveratrol also lowers Abeta levels in cell lines; however, the underlying mechanism responsible for this effect is largely unknown. Moreover, the bioavailability of resveratrol in the brain remains uncertain. Here we show that AMPK signaling controls Abeta metabolism and mediates the anti-amyloidogenic effect of resveratrol in non-neuronal and neuronal cells, including in mouse primary neurons. Resveratrol increased cytosolic calcium levels and promoted AMPK activation by the calcium/calmodulin-dependent protein kinase kinase-beta. Direct pharmacological and genetic activation of AMPK lowered extracellular Abeta accumulation, whereas AMPK inhibition reduced the effect of resveratrol on Abeta levels. Furthermore, resveratrol inhibited the AMPK target mTOR (mammalian target of rapamycin) to trigger autophagy and lysosomal degradation of Abeta. Finally, orally administered resveratrol in mice was detected in the brain where it activated AMPK and reduced cerebral Abeta levels and deposition in the cortex. These data suggest that resveratrol and pharmacological activation of AMPK have therapeutic potential against Alzheimer disease.
Collapse
Affiliation(s)
- Valérie Vingtdeux
- From the Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030
| | - Luca Giliberto
- From the Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030
| | - Haitian Zhao
- From the Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030
| | - Pallavi Chandakkar
- From the Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030
| | - Qingli Wu
- the New Use Agriculture and Natural Plant Products Program, Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901
| | - James E. Simon
- the New Use Agriculture and Natural Plant Products Program, Department of Plant Biology and Pathology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901
| | | | - Jessica Lobo
- Food Science, Purdue University, West Lafayette, Indiana 47907, and
| | - Mario G. Ferruzzi
- the Departments of Foods and Nutrition and
- Food Science, Purdue University, West Lafayette, Indiana 47907, and
| | - Peter Davies
- From the Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030
- the Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Philippe Marambaud
- From the Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030
- the Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
114
|
Structure and biochemical analysis of the heparin-induced E1 dimer of the amyloid precursor protein. Proc Natl Acad Sci U S A 2010; 107:5381-6. [PMID: 20212142 DOI: 10.1073/pnas.0911326107] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The amyloid precursor protein (APP) is the key player in Alzheimer's disease pathology, yet APP and its analogues are also essential for neuronal development and cell homeostasis in mammals. We have determined the crystal structure of the entire N-terminal APP-E1 domain consisting of the growth factor like and the copper binding domains at 2.7-A resolution and show that E1 functions as a rigid functional entity. The two subdomains interact tightly in a pH-dependent manner via an evolutionarily conserved interface area. Two E1 entities dimerize upon their interaction with heparin, requiring 8-12 sugar rings to form the heparin-bridged APP-E1 dimer in an endothermic and pH-dependent process that is characterized by a low micromolar dissociation constant. Limited proteolysis confirms that the heparin-bridged E1 dimers obtained in solution correspond to a dimer contact in our crystal, enabling us to model this heparin-[APP-E1](2) complex. Correspondingly, the APP-based signal transduction, cell-cell- and/or cell-ECM interaction should depend on dimerization induced by heparin, as well as on pH, arguing that APP could fulfill different functions depending on its (sub)cellular localization.
Collapse
|
115
|
Virata MJ, Zeller RW. Ascidians: an invertebrate chordate model to study Alzheimer's disease pathogenesis. Dis Model Mech 2010; 3:377-85. [PMID: 20197417 DOI: 10.1242/dmm.003434] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Here we present the ascidian Ciona intestinalis as an alternative invertebrate system to study Alzheimer's disease (AD) pathogenesis. Through the use of AD animal models, researchers often attempt to reproduce various aspects of the disease, particularly the coordinated processing of the amyloid precursor protein (APP) by alpha-, beta- and gamma-secretases to generate amyloid beta (Abeta)-containing plaques. Recently, Drosophila and C. elegans AD models have been developed, exploiting the relative simplicity of these invertebrate systems, but they lack a functional Abeta sequence and a beta-secretase ortholog, thus complicating efforts to examine APP processing in vivo. We propose that the ascidian is a more appropriate invertebrate AD model owing to their phylogenetic relationship with humans. This is supported by bioinformatic analyses, which indicate that the ascidian genome contains orthologs of all AD-relevant genes. We report that transgenic ascidian larvae can properly process human APP(695) to generate Abeta peptides. Furthermore, Abeta can rapidly aggregate to form amyloid-like plaques, and plaque deposition is significantly increased in larvae expressing a human APP(695) variant associated with familial Alzheimer's disease. We also demonstrate that nervous system-specific Abeta expression alters normal larval behavior during attachment. Importantly, plaque formation and alterations in behavior are not only observed within 24 hours post-fertilization, but anti-amyloid drug treatment improves these AD-like pathologies. This ascidian model for AD provides a powerful and rapid system to study APP processing, Abeta plaque formation and behavioral alterations, and could aid in identifying factors that modulate amyloid deposition and the associated disruption of normal cellular function and behaviors.
Collapse
Affiliation(s)
- Michael J Virata
- Center for Applied and Experimental Genomics, San Diego State University, 5500 Campanile Drive, San Diego, California 92182-4614, USA
| | | |
Collapse
|
116
|
Lyubartseva G, Smith JL, Markesbery WR, Lovell MA. Alterations of zinc transporter proteins ZnT-1, ZnT-4 and ZnT-6 in preclinical Alzheimer's disease brain. Brain Pathol 2010; 20:343-50. [PMID: 19371353 PMCID: PMC3175637 DOI: 10.1111/j.1750-3639.2009.00283.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 02/04/2009] [Accepted: 02/09/2009] [Indexed: 12/01/2022] Open
Abstract
Our previous studies demonstrate alterations of zinc (Zn) transporter proteins ZnT-1, ZnT-4 and ZnT-6 in vulnerable brain regions of subjects with mild cognitive impairment (MCI), and early and late stage Alzheimer's disease (AD), suggesting disruptions of Zn homeostasis may play a role in the pathogenesis of AD. A preclinical stage of AD (PCAD) has been described in which subjects show no overt clinical manifestations of AD, but demonstrate significant AD pathology at autopsy. To determine if alterations of ZnT proteins occur in PCAD, we measured ZnT-1, ZnT-4 and ZnT-6 in the hippocampus/parahippocampal gyrus (HPG) and cerebellum (CER) of seven PCAD subjects and seven age-matched normal control (NC) subjects using Western blot analysis and immunohistochemistry. Our results show a significant decrease (P < 0.05) of ZnT-1 in HPG of PCAD subjects, along with an increase of ZnT-4 in PCAD CER and ZnT-6 in PCAD HPG, but a significant decrease in PCAD CER compared to NC subjects. Confocal microscopy of representative sections of HPG shows altered ZnTs are associated with neurons immunopositive for MC-1, a monoclonal antibody that identifies neurons early in formation of neurofibrillary tangles. Overall, our results suggest that alterations in Zn transport proteins may contribute to the pathology observed in PCAD subjects before onset of clinical symptoms.
Collapse
Affiliation(s)
| | | | - William R. Markesbery
- Alzheimer's Disease Center, Sanders‐Brown Center on Aging
- Departments of Neurology and Pathology, University of Kentucky, Lexington, Ky
| | - Mark A. Lovell
- Department of Chemistry
- Alzheimer's Disease Center, Sanders‐Brown Center on Aging
| |
Collapse
|
117
|
Early effects of aluminum chloride on beta-secretase mRNA expression in a neuronal model of beta-amyloid toxicity. Cell Biol Toxicol 2010; 26:367-77. [PMID: 20111991 DOI: 10.1007/s10565-009-9149-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Accepted: 12/20/2009] [Indexed: 12/25/2022]
Abstract
Amyloid beta peptide (Abeta), generated by proteolytic cleavage of the amyloid precursor protein (APP), plays a pivotal role in the pathogenesis of Alzheimer's disease (AD). The key step in the generation of Abeta is cleavage of APP by beta-secretases (beta-site APP-cleaving enzyme 1 (BACE1) and BACE2). There has been suggestion of interaction between aluminum and several AD-associated pathways. However, the underlying mechanisms still remain unclear. Here, we report the effects of aluminum chloride (AlCl(3)) in Abeta-induced toxicity using differentiated neuronal SH-SY5Y cells. The metal significantly enhances Abeta-induced cell death at concentrations ranging from 50 to 300 microM after 24 and 48 h. After 72 and 96 h treatment, cell death is increased already at 10 microM. Early coexposure of cells to 10 microM AlCl(3) and 2 microM Abeta differentially affected beta-secretase mRNA levels as compared to single Abeta treatment after 1 and 3 h. BACE1 levels were slightly reduced after 1 h and significantly increased after 3 h exposure, whereas BACE2 levels were increased at both times considered. Both genes' mRNA levels were downregulated at longer times (6, 12, and 24 h). Although these results indicate that aluminum toxicity is correlated to changes in both BACE1 and BACE2 expression levels, the subsequent common downregulation observed suggests that aluminum involvement in the Abeta cascade is subtle, and other underlying mechanisms might be involved.
Collapse
|
118
|
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: 70] [Impact Index Per Article: 5.0] [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
|
119
|
Bakshi P, Jin C, Broutin P, Berhane B, Reed J, Mullan M. Structural optimization of a CXCR2-directed antagonist that indirectly inhibits γ-secretase and reduces Aβ. Bioorg Med Chem 2009; 17:8102-12. [DOI: 10.1016/j.bmc.2009.09.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/17/2009] [Accepted: 09/30/2009] [Indexed: 01/13/2023]
|
120
|
Perez JL, Carrero I, Gonzalo P, Arevalo-Serrano J, Sanz-Anquela JM, Ortega J, Rodriguez M, Gonzalo-Ruiz A. Soluble oligomeric forms of beta-amyloid (Abeta) peptide stimulate Abeta production via astrogliosis in the rat brain. Exp Neurol 2009; 223:410-21. [PMID: 19879263 DOI: 10.1016/j.expneurol.2009.10.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 09/29/2009] [Accepted: 10/17/2009] [Indexed: 10/20/2022]
Abstract
The aim of this study was to investigate the interaction between beta-amyloid (Abeta) peptide and astrogliosis in early stages of Abeta toxicity. In Wistar rats, anaesthetised with equitesine, a single microinjection of Abeta1-42 oligomers was placed into the retrosplenial cortex. Control animals were injected with Abeta42-1 peptide into the corresponding regions of cerebral cortex. Immunocytochemical analysis revealed an intense Abeta immunoreactivity (IR) at the level of Abeta1-42 injection site, increasing from the first 24 h to later (72 h) time point. Control injection showed a light staining surrounding the injection site. In Abeta oligomers-treated animals, Abeta-immunopositive product also accumulates in cortical cells, particularly in frontal and temporal cortices at an early (24 h) time point. Abeta-IR structures-like diffuse aggregates forms were also observed in hippocampus and in several cortical areas, increasing from the first 24 h to later (72 h) time point. In control animals no specific staining was seen neither in cortical cells nor in structures-like diffuse aggregates forms. Injections of Abeta oligomers also induce activation of astrocytes surrounding and infiltrating the injection site. Astrocyte activation is evidenced by morphological changes and upregulation of glial fibrillary acidic protein (GFAP). By GFAP immunoblotting we detected two immunopositive protein bands, at 50 and 48 kDa molecular mass. Confocal analysis also showed that GFAP co-localized with Abeta-IR material in a time-dependent manner. In conclusion, our results indicate that astrocyte activation might have a critical role in the mechanisms of Abeta-induced neurodegeneration, and that should be further studied as possible targets for therapeutic intervention in AD.
Collapse
Affiliation(s)
- J L Perez
- Laboratory of Neuroanatomy, Institute of Neuroscience of Castilla and León, Valladolid University (Campus of Soria), Spain
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Matsuda S, Matsuda Y, Snapp EL, D'Adamio L. Maturation of BRI2 generates a specific inhibitor that reduces APP processing at the plasma membrane and in endocytic vesicles. Neurobiol Aging 2009; 32:1400-8. [PMID: 19748705 DOI: 10.1016/j.neurobiolaging.2009.08.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 07/28/2009] [Accepted: 08/17/2009] [Indexed: 11/18/2022]
Abstract
Processing of the amyloid-β (Aβ) precursor protein (APP) has been extensively studied since it leads to production of Aβ peptides. Toxic forms of Aβ aggregates are considered the cause of Alzheimer's disease (AD). On the other end, BRI2 is implicated in APP processing and Aβ production. We have investigated the precise mechanism by which BRI2 modulates APP cleavages and have found that BRI2 forms a mature BRI2 polypeptide that is transported to the plasma membrane and endosomes where it interacts with mature APP. Notably, immature forms of APP and BRI2 fail to interact. Mature BRI2 inhibits APP processing by α-, β- and γ-secretases on the plasma membrane and in endocytic compartments. Thus, BRI2 is a specific inhibitor that reduces secretases' access to APP in the intracellular compartments where APP is normally processed.
Collapse
Affiliation(s)
- Shuji Matsuda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
122
|
|
123
|
Synaptic NMDA receptor activation stimulates alpha-secretase amyloid precursor protein processing and inhibits amyloid-beta production. J Neurosci 2009; 29:4442-60. [PMID: 19357271 DOI: 10.1523/jneurosci.6017-08.2009] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Altered amyloid precursor protein (APP) processing leading to increased production and oligomerization of Abeta may contribute to Alzheimer's disease (AD). Understanding how APP processing is regulated under physiological conditions may provide new insights into AD pathogenesis. Recent reports demonstrate that excitatory neural activity regulates APP metabolism and Abeta levels, although understanding of the molecular mechanisms involved is incomplete. We have investigated whether NMDA receptor activity regulates APP metabolism in primary cultured cortical neurons. We report that a pool of APP is localized to the postsynaptic compartment in cortical neurons and observed partial overlap of APP with both NR1 and PSD-95. NMDA receptor stimulation increased nonamyloidogenic alpha-secretase-mediated APP processing, as measured by a 2.5-fold increase in cellular alpha-C-terminal fragment (C83) levels after glutamate or NMDA treatment. This increase was blocked by the NMDA receptor antagonists d-AP5 and MK801 but not by the AMPA receptor antagonist CNQX or the L-type calcium channel blocker nifedipine, was prevented by chelation of extracellular calcium, and was blocked by the alpha-secretase inhibitor TAPI-1. Cotreatment of cortical neurons with bicuculline and 4-AP, which stimulates glutamate release and activates synaptic NMDA receptors, evoked an MK801-sensitive increase in C83 levels. Furthermore, NMDA receptor stimulation caused a twofold increase in the amount of soluble APP detected in the neuronal culture medium. Finally, NMDA receptor activity inhibited both Abeta1-40 release and Gal4-dependent luciferase activity induced by beta-gamma-secretase-mediated cleavage of an APP-Gal4 fusion protein. Altogether, these data suggest that calcium influx through synaptic NMDA receptors promotes nonamyloidogenic alpha-secretase-mediated APP processing.
Collapse
|
124
|
Marambaud P, Dreses-Werringloer U, Vingtdeux V. Calcium signaling in neurodegeneration. Mol Neurodegener 2009; 4:20. [PMID: 19419557 PMCID: PMC2689218 DOI: 10.1186/1750-1326-4-20] [Citation(s) in RCA: 232] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 05/06/2009] [Indexed: 12/16/2022] Open
Abstract
Calcium is a key signaling ion involved in many different intracellular and extracellular processes ranging from synaptic activity to cell-cell communication and adhesion. The exact definition at the molecular level of the versatility of this ion has made overwhelming progress in the past several years and has been extensively reviewed. In the brain, calcium is fundamental in the control of synaptic activity and memory formation, a process that leads to the activation of specific calcium-dependent signal transduction pathways and implicates key protein effectors, such as CaMKs, MAPK/ERKs, and CREB. Properly controlled homeostasis of calcium signaling not only supports normal brain physiology but also maintains neuronal integrity and long-term cell survival. Emerging knowledge indicates that calcium homeostasis is not only critical for cell physiology and health, but also, when deregulated, can lead to neurodegeneration via complex and diverse mechanisms involved in selective neuronal impairments and death. The identification of several modulators of calcium homeostasis, such as presenilins and CALHM1, as potential factors involved in the pathogenesis of Alzheimer's disease, provides strong support for a role of calcium in neurodegeneration. These observations represent an important step towards understanding the molecular mechanisms of calcium signaling disturbances observed in different brain diseases such as Alzheimer's, Parkinson's, and Huntington's diseases.
Collapse
Affiliation(s)
- Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, New York 11030, USA.
| | | | | |
Collapse
|
125
|
Perdivara I, Petrovich R, Allinquant B, Alliquant B, Deterding LJ, Tomer KB, Przybylski M. Elucidation of O-glycosylation structures of the beta-amyloid precursor protein by liquid chromatography-mass spectrometry using electron transfer dissociation and collision induced dissociation. J Proteome Res 2009; 8:631-42. [PMID: 19093876 DOI: 10.1021/pr800758g] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Accumulation and deposition of beta-amyloid peptide, a major constituent in neuritic plaques are hallmarks of Alzheimer's disease (AD) and AD-related neurodegenerative diseases. beta-Amyloid (Abeta) is derived from the proteolytic cleavage of amyloid precursor protein (APP), a transmembrane protein present in three major isoforms in brain comprising 695, 751 and 770 amino acids, respectively. Among other post-translational modifications, APP is modified during maturation by N- and O-glycosylation, which are thought to be responsible for its expression and secretion. Unlike N-glycosylation, no sites of O-glycosylation of APP have previously been reported. We report here the identification of three specific O-glycosylation sites of the secreted APP695 (sAPP695) produced in CHO cells, using a combination of high-performance liquid chromatography and electrospray-tandem mass spectrometry. With the use of electron transfer dissociation and collision induced dissociation (ETD and CID), we identified type, composition and structures of the Core 1 type O-linked glycans attached at the residues Thr 291, Thr 292 and Thr 576 of the full-length APP695. The glycosylations comprise multiple short glycans, containing N-acetyl galactosamine (GalNAc), Gal-GalNAc and sialic acid terminated structures. The presence of the glycopeptides in the tryptic mixture was identified using the CID-generated sugar oxonium ions. ETD proved to be valuable for the unambiguous identification of the modified sites as ETD fragmentation occurred along the peptide backbone with little or no cleavage of the glycans. Thus, the combination of the CID and ETD techniques in LC-MS is shown here, as a powerful tool for de novo identification of O-glycosylations at unknown modification sites in proteins.
Collapse
Affiliation(s)
- Irina Perdivara
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | |
Collapse
|
126
|
Mehta TK, Dougherty JJ, Wu J, Choi CH, Khan GM, Nichols RA. Defining pre-synaptic nicotinic receptors regulated by beta amyloid in mouse cortex and hippocampus with receptor null mutants. J Neurochem 2009; 109:1452-8. [PMID: 19457164 DOI: 10.1111/j.1471-4159.2009.06070.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Disruption of neuronal signaling by soluble beta-amyloid has been implicated in deficits in short-term recall in the early stages of Alzheimer's disease. One potential target for beta-amyloid is the synapse, with evidence for differential interaction with both pre- and post-synaptic elements. Our previous work revealed an agonist-like action of soluble beta-amyloid (pM to nM) on isolated pre-synaptic terminals to increase [Ca(2+)]i, with apparent involvement of pre-synaptic nicotinic receptors. To directly establish the role of nicotinic receptors in pre-synaptic Ca(2+) regulation, we investigated the pre-synaptic action of beta-amyloid on terminals isolated from mice harboring either beta2 or alpha7 nicotinic receptor null mutants (knockouts). Average pre-synaptic responses to beta-amyloid in hippocampal terminals of alpha7 knockout mice were unchanged, whereas responses in hippocampal terminals from beta2 knockout mice were strongly attenuated. In contrast, pre-synaptic responses to soluble beta-amyloid were strongly attenuated in cortical terminals from alpha7 knockout mice but were moderately attenuated in cortical terminals from beta2 knockout mice. The latter responses, having distinct kinetics, were completely blocked by alpha-bungarotoxin. The use of receptor null mutants thus permitted direct demonstration of the involvement of specific nicotinic receptors in pre-synaptic Ca(2+) regulation by soluble beta-amyloid, and also indicated differential neuromodulation by beta-amyloid of synapses in hippocampus and cortex.
Collapse
Affiliation(s)
- Tejal K Mehta
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
127
|
Castellani RJ, Zhu X, Lee HG, Smith MA, Perry G. Molecular pathogenesis of Alzheimer's disease: reductionist versus expansionist approaches. Int J Mol Sci 2009; 10:1386-1406. [PMID: 19399255 PMCID: PMC2672036 DOI: 10.3390/ijms10031386] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 03/20/2009] [Accepted: 03/23/2009] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is characterized clinically by dementia and pathologically by two hallmark lesions, senile plaques and neurofibrillary tangles. About a quarter century ago these hallmark lesions were purified and their protein constituents identified, precipitating an avalanche of molecular studies as well as substantial optimism about successful therapeutic intervention. In 2009, we now have copious knowledge on the biochemical cascades that produce these proteins, the different modifications and forms in which these proteins exist, and the ability to selectively target these proteins for therapeutic intervention on an experimental basis. At the same time, there has been no discernible alteration in the natural course of AD in humans. While it may be that the complexity of AD will exceed our capacity to make significant treatment progress for decades or more, a paradigm shift from the reductionism that defines amyloid-beta and tau hypotheses, to one that more accurately reflects the meaning of neuropathological changes, may be warranted. We and others have demonstrated that AD pathology is a manifestation of cellular adaptation, specifically as a defense against oxidative injury. As such, AD pathology is therefore a host response rather than a manifestation of cytotoxic protein injury, and is unlikely to be a fruitful target for therapeutic intervention. An "expansionist" view of the disease, we believe, with oxidative stress as a pleiotropic and upstream process, more aptly describes the relationship between various and numerous molecular alterations and clinical disease.
Collapse
Affiliation(s)
- Rudy J. Castellani
- Division of Neuropathology, University of Maryland, Baltimore, Maryland, USA
- Author to whom correspondence should be addressed; E-Mail:
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hyoung-Gon Lee
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark A. Smith
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - George Perry
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- College of Sciences, University of Texas, San Antonio, Texas, USA
| |
Collapse
|
128
|
Thathiah A, Spittaels K, Hoffmann M, Staes M, Cohen A, Horré K, Vanbrabant M, Coun F, Baekelandt V, Delacourte A, Fischer DF, Pollet D, De Strooper B, Merchiers P. The orphan G protein-coupled receptor 3 modulates amyloid-beta peptide generation in neurons. Science 2009; 323:946-51. [PMID: 19213921 DOI: 10.1126/science.1160649] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Deposition of the amyloid-beta peptide is a pathological hallmark of Alzheimer's disease. A high-throughput functional genomics screen identified G protein-coupled receptor 3 (GPR3), a constitutively active orphan G protein-coupled receptor, as a modulator of amyloid-beta production. Overexpression of GPR3 stimulated amyloid-beta production, whereas genetic ablation of GPR3 prevented accumulation of the amyloid-beta peptide in vitro and in an Alzheimer's disease mouse model. GPR3 expression led to increased formation and cell-surface localization of the mature gamma-secretase complex in the absence of an effect on Notch processing. GPR3 is highly expressed in areas of the normal human brain implicated in Alzheimer's disease and is elevated in the sporadic Alzheimer's disease brain. Thus, GPR3 represents a potential therapeutic target for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Amantha Thathiah
- Department of Molecular and Developmental Genetics, Vlaams Institute for Biotechnology, Center for Human Genetics, Catholic University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Souplet V, Desmet R, Melnyk O. In Situ Ligation between Peptides and Silica Nanoparticles for Making Peptide Microarrays on Polycarbonate. Bioconjug Chem 2009; 20:550-7. [DOI: 10.1021/bc800474g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vianney Souplet
- Institut de Biologie de Lille, UMR CNRS 8161, Universités de Lille 1 et 2, Institut Pasteur de Lille, IFR 142, 1 rue du Professeur Calmette, 59021 Lille Cedex, France
| | - Rémi Desmet
- Institut de Biologie de Lille, UMR CNRS 8161, Universités de Lille 1 et 2, Institut Pasteur de Lille, IFR 142, 1 rue du Professeur Calmette, 59021 Lille Cedex, France
| | - Oleg Melnyk
- Institut de Biologie de Lille, UMR CNRS 8161, Universités de Lille 1 et 2, Institut Pasteur de Lille, IFR 142, 1 rue du Professeur Calmette, 59021 Lille Cedex, France
| |
Collapse
|
130
|
Tang BL. Neuronal protein trafficking associated with Alzheimer disease: from APP and BACE1 to glutamate receptors. Cell Adh Migr 2009; 3:118-28. [PMID: 19372755 DOI: 10.4161/cam.3.1.7254] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aberrant and/or cumulative amyloid-beta (Abeta) production, resulting from proteolytic processing of the amyloid precursor protein (APP) by beta and gamma-secretases, have been postulated to be a main etiological basis of Alzheimer disease (AD). A number of proteins influence the subcellular trafficking itinerary of APP and the beta-site APP-cleaving enzyme (BACE1) between the cell surface, endosomes and the trans-Golgi network (TGN). Available evidence suggests that co-residence of APP and BACE1 in the endosomal compartments promotes amyloidogenesis. Retrograde transport of APP out of the endosome to the TGN reduces Abeta production, while APP routed to and kept at the cell surface enhances its non-amyloidogenic, alpha-secretase-mediated processing. Changes in post-Golgi membrane trafficking in aging neurons that may influence APP processing is particularly relevant to late-onset, idiopathic AD. Dystrophic axons are key features of AD pathology, and impaired axonal transport could play crucial roles in the pathogenesis of idiopathic AD. Recent evidence has also indicated that Abeta-induced synaptic defects and memory impairment could be explained by a loss of both AMPA and NMDA receptors through endocytosis. Detail understanding of factors that influence these neuronal trafficking processes will open up novel therapeutic avenues for preventing or delaying the onset of symptomatic AD.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
131
|
Lovell MA. A potential role for alterations of zinc and zinc transport proteins in the progression of Alzheimer's disease. J Alzheimers Dis 2009; 16:471-83. [PMID: 19276540 PMCID: PMC2881701 DOI: 10.3233/jad-2009-0992] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although multiple studies have suggested a role for alterations of zinc (Zn) and zinc transport (ZnT) proteins in the pathogenesis of Alzheimer's disease, the exact role of this essential trace element in the progression of the disease remains unclear. The following review discusses the normal role of Zn and ZnT proteins in brain and the potential effects of their alteration in the pathogenesis of Alzheimer's disease, particularly in the processing of the amyloid-beta protein precursor and amyloid-beta peptide generation and aggregation.
Collapse
Affiliation(s)
- Mark A Lovell
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
132
|
Zhang L, Yu H, Song C, Lin X, Chen B, Tan C, Cao G, Wang Z. Expression, purification, and characterization of recombinant human beta-amyloid42 peptide in Escherichia coli. Protein Expr Purif 2008; 64:55-62. [PMID: 18996484 DOI: 10.1016/j.pep.2008.10.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 10/10/2008] [Accepted: 10/10/2008] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive function. Evidence indicates that abnormal processing and extracellular deposition of the beta-amyloid42 peptide, the longer form of proteolytic derivative of the transmembrane glycoprotein-amyloid precursor protein (APP), is a key step in the pathogenesis of AD. Since it is convenient and economical to obtain such a peptide biologically, in this study, we report for the first time a method to express in E. coli and purify beta-amyloid42 using glutathione-S-transferase (GST) fusion system. beta-Amyloid42 gene was inserted into a vector pGEX-4T-1 to construct a GST-fusion protein. The fusion protein GST-beta-amyloid42, expressed in BL21 (DE3) strain, was purified with GSH-affinity chromatography followed by thrombin cleavage. The digested product was further purified with an additional GSH-affinity and a Benzamidine chromatography step. After cleavage and purification, the beta-amyloid42 moiety showed the expected size of 4.5 kDa on Tricine-SDS-PAGE, and was further confirmed by Western blot. Moreover, the fibrillar recombinant beta-amyloid42 exhibited great aggregation activity and showed neurotoxicity on neuron cells in vitro. These results suggest that our method will be useful in obtaining a large quantity of recombinant beta-amyloid42 peptide for further physiological and biochemical studies.
Collapse
Affiliation(s)
- Li Zhang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu 214063, China
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Schmidt C, Lepsverdize E, Chi SL, Das AM, Pizzo SV, Dityatev A, Schachner M. Amyloid precursor protein and amyloid beta-peptide bind to ATP synthase and regulate its activity at the surface of neural cells. Mol Psychiatry 2008; 13:953-69. [PMID: 17726461 DOI: 10.1038/sj.mp.4002077] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Amyloid precursor protein (APP) and amyloid beta-peptide (Abeta) have been implicated in a variety of physiological and pathological processes underlying nervous system functions. APP shares many features with adhesion molecules in that it is involved in neurite outgrowth, neuronal survival and synaptic plasticity. It is, thus, of interest to identify binding partners of APP that influence its functions. Using biochemical cross-linking techniques we have identified ATP synthase subunit alpha as a binding partner of the extracellular domain of APP and Abeta. APP and ATP synthase colocalize at the cell surface of cultured hippocampal neurons and astrocytes. ATP synthase subunit alpha reaches the cell surface via the secretory pathway and is N-glycosylated during this process. Transfection of APP-deficient neuroblastoma cells with APP results in increased surface localization of ATP synthase subunit alpha. The extracellular domain of APP and Abeta partially inhibit the extracellular generation of ATP by the ATP synthase complex. Interestingly, the binding sequence of APP and Abeta is similar in structure to the ATP synthase-binding sequence of the inhibitor of F1 (IF(1)), a naturally occurring inhibitor of the ATP synthase complex in mitochondria. In hippocampal slices, Abeta and IF(1) similarly impair both short- and long-term potentiation via a mechanism that could be suppressed by blockade of GABAergic transmission. These observations indicate that APP and Abeta regulate extracellular ATP levels in the brain, thus suggesting a novel mechanism in Abeta-mediated Alzheimer's disease pathology.
Collapse
Affiliation(s)
- C Schmidt
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
134
|
De Vos KJ, Grierson AJ, Ackerley S, Miller CCJ. Role of axonal transport in neurodegenerative diseases. Annu Rev Neurosci 2008; 31:151-73. [PMID: 18558852 DOI: 10.1146/annurev.neuro.31.061307.090711] [Citation(s) in RCA: 522] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many major human neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS), display axonal pathologies including abnormal accumulations of proteins and organelles. Such pathologies highlight damage to the axon as part of the pathogenic process and, in particular, damage to transport of cargoes through axons. Indeed, we now know that disruption of axonal transport is an early and perhaps causative event in many of these diseases. Here, we review the role of axonal transport in neurodegenerative disease.
Collapse
Affiliation(s)
- Kurt J De Vos
- MRC Center for Neurodegeneration Research, Institute of Psychiatry, King's College, London SE5 8AF, United Kingdom.
| | | | | | | |
Collapse
|
135
|
Wahlström A, Hugonin L, Perálvarez-Marín A, Jarvet J, Gräslund A. Secondary structure conversions of Alzheimer's Abeta(1-40) peptide induced by membrane-mimicking detergents. FEBS J 2008; 275:5117-28. [PMID: 18786140 DOI: 10.1111/j.1742-4658.2008.06643.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The amyloid beta peptide (Abeta) with 39-42 residues is the major component of amyloid plaques found in brains of Alzheimer's disease patients, and soluble oligomeric peptide aggregates mediate toxic effects on neurons. The Abeta aggregation involves a conformational change of the peptide structure to beta-sheet. In the present study, we report on the effect of detergents on the structure transitions of Abeta, to mimic the effects that biomembranes may have. In vitro, monomeric Abeta(1-40) in a dilute aqueous solution is weakly structured. By gradually adding small amounts of sodium dodecyl sulfate (SDS) or lithium dodecyl sulfate to a dilute aqueous solution, Abeta(1-40) is converted to beta-sheet, as observed by CD at 3 degrees C and 20 degrees C. The transition is mainly a two-state process, as revealed by approximately isodichroic points in the titrations. Abeta(1-40) loses almost all NMR signals at dodecyl sulfate concentrations giving rise to the optimal beta-sheet content (approximate detergent/peptide ratio = 20). Under these conditions, thioflavin T fluorescence measurements indicate a maximum of aggregated amyloid-like structures. The loss of NMR signals suggests that these are also involved in intermediate chemical exchange. Transverse relaxation optimized spectroscopy NMR spectra indicate that the C-terminal residues are more dynamic than the others. By further addition of SDS or lithium dodecyl sulfate reaching concentrations close to the critical micellar concentration, CD, NMR and FTIR spectra show that the peptide rearranges to form a micelle-bound structure with alpha-helical segments, similar to the secondary structures formed when a high concentration of detergent is added directly to the peptide solution.
Collapse
Affiliation(s)
- Anna Wahlström
- Department of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural Sciences, Stockholm University, Sweden
| | | | | | | | | |
Collapse
|
136
|
Dreses-Werringloer U, Lambert JC, Vingtdeux V, Zhao H, Vais H, Siebert A, Jain A, Koppel J, Rovelet-Lecrux A, Hannequin D, Pasquier F, Galimberti D, Scarpini E, Mann D, Lendon C, Campion D, Amouyel P, Davies P, Foskett JK, Campagne F, Marambaud P. A polymorphism in CALHM1 influences Ca2+ homeostasis, Abeta levels, and Alzheimer's disease risk. Cell 2008; 133:1149-61. [PMID: 18585350 DOI: 10.1016/j.cell.2008.05.048] [Citation(s) in RCA: 252] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/30/2008] [Accepted: 05/22/2008] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a genetically heterogeneous disorder characterized by early hippocampal atrophy and cerebral amyloid-beta (Abeta) peptide deposition. Using TissueInfo to screen for genes preferentially expressed in the hippocampus and located in AD linkage regions, we identified a gene on 10q24.33 that we call CALHM1. We show that CALHM1 encodes a multipass transmembrane glycoprotein that controls cytosolic Ca(2+) concentrations and Abeta levels. CALHM1 homomultimerizes, shares strong sequence similarities with the selectivity filter of the NMDA receptor, and generates a large Ca(2+) conductance across the plasma membrane. Importantly, we determined that the CALHM1 P86L polymorphism (rs2986017) is significantly associated with AD in independent case-control studies of 3404 participants (allele-specific OR = 1.44, p = 2 x 10(-10)). We further found that the P86L polymorphism increases Abeta levels by interfering with CALHM1-mediated Ca(2+) permeability. We propose that CALHM1 encodes an essential component of a previously uncharacterized cerebral Ca(2+) channel that controls Abeta levels and susceptibility to late-onset AD.
Collapse
Affiliation(s)
- Ute Dreses-Werringloer
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
|
138
|
Lack of pathology in a triple transgenic mouse model of Alzheimer's disease after overexpression of the anti-apoptotic protein Bcl-2. J Neurosci 2008; 28:3051-9. [PMID: 18354008 DOI: 10.1523/jneurosci.5620-07.2008] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of plaques containing beta-amyloid (Abeta) and neurofibrillary tangles (NFTs) consisting of modified tau. Although Abeta deposition is thought to precede the formation of NFTs in AD, the molecular steps connecting these two pathologies is not known. Previous studies have suggested that caspase activation plays an important role in promoting the pathology associated with AD. To further understand the contribution of caspases in disease progression, a triple transgenic Alzheimer's mouse model overexpressing the anti-apoptotic protein Bcl-2 was generated. Here we show that overexpression of Bcl-2 limited caspase-9 activation and reduced the caspase cleavage of tau. Moreover, overexpression of Bcl-2 attenuated the processing of APP (amyloid precursor protein) and tau and reduced the number of NFTs and extracellular deposits of Abeta associated with these animals. In addition, overexpression of Bcl-2 in 3xTg-AD mice improved place recognition memory. These findings suggest that the activation of apoptotic pathways may be an early event in AD and contributes to the pathological processes that promote the disease mechanisms underlying AD.
Collapse
|
139
|
Patey S, Edwards E, Yates E, Turnbull J. Engineered Heparins: Novel β-Secretase Inhibitors as Potential Alzheimer’s Disease Therapeutics. NEURODEGENER DIS 2008; 5:197-9. [DOI: 10.1159/000113701] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
140
|
He J, Luo H, Yan B, Yu Y, Wang H, Wei Z, Zhang Y, Xu H, Tempier A, Li X, Li XM. Beneficial effects of quetiapine in a transgenic mouse model of Alzheimer's disease. Neurobiol Aging 2008; 30:1205-16. [PMID: 18079026 DOI: 10.1016/j.neurobiolaging.2007.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 10/11/2007] [Accepted: 11/02/2007] [Indexed: 11/17/2022]
Abstract
Previous studies have suggested that quetiapine, an atypical antipsychotic drug, may have beneficial effects on cognitive impairment, and be a neuroprotectant in treating neurodegenerative diseases. In the present study, we investigated the effects of quetiapine on memory impairment and pathological changes in an amyloid precursor protein (APP)/presenilin-1 (PS-1) double transgenic mouse model of Alzheimer's disease (AD). Non-transgenic and transgenic mice were treated with quetiapine (0, 2.5, or 5mg/(kg day)) for 1, 4, and 7 months in drinking water from the age of 2 months. After 4 and 7 months of continuous quetiapine administration, memory impairment was prevented, and the number of beta-amyloid (Abeta) plaques decreased in the cortex and hippocampus of the transgenic mice. Quetiapine also decreased brain Abeta peptides, beta-secretase activity and expression, and the level of C99 (an APP C-terminal fragment following cleavage by beta-secretase) in the transgenic mice. Furthermore, quetiapine attenuated anxiety-like behavior, up-regulated cerebral Bcl-2 protein, and decreased cerebral nitrotyrosine in the transgenic mice. These findings suggest that quetiapine can alleviate cognitive impairment and pathological changes in an APP/PS1 double transgenic mouse model of AD, and further indicate that quetiapine may have preventive effects in the treatment of AD.
Collapse
Affiliation(s)
- Jue He
- Laboratory of Neuropharmacology, Wenzhou Medical College, Wenzhou, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Lewis V, Collins SJ. Analysis of endogenous PrPC processing in neuronal and non-neuronal cell lines. Methods Mol Biol 2008; 459:229-239. [PMID: 18576158 DOI: 10.1007/978-1-59745-234-2_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Numerous transmembrane and glycosylphosphatidylinositol (GPI)-anchored proteins, covering a vast range of structural and functional classes, are recognized to undergo proteolytic cleavage or shedding from the plasma membrane. Although this widespread phenomenon seems fundamental to normal cellular biology, proteolytic processing also seems to play a central role in the pathogenesis of some neurodegenerative disorders such as Alzheimer's disease. An analogous situation may exist in prion disorders. The GPI-anchored cellular prion protein (PrP(C)) may be endoproteolytically cleaved at two different sites: one at the C-terminal end of the octameric repeat region and the other within a potentially neurotoxic and amyloidogenic region of the protein. The relevance of these alternative proteolytic events to normal cell function and pathogenesis is incompletely resolved. Study and characterization of the constitutive processing of PrP(C) will provide insight into the biological relevance of alternative cleavages in terms of normal PrP(C) function, and also into the potential role, if any, to disease causation.
Collapse
Affiliation(s)
- Victoria Lewis
- Australian National Creutzfeldt-Jakob Disease Registry, Department of Pathology, University of Melbourne, Melbourne, Australia
| | | |
Collapse
|
142
|
Cai L, Liow JS, Zoghbi SS, Cuevas J, Baetas C, Hong J, Shetty HU, Seneca NM, Brown AK, Gladding R, Temme SS, Herman MM, Innis RB, Pike VW. Synthesis and Evaluation of N-Methyl and S-Methyl 11C-Labeled 6-Methylthio-2-(4′-N,N-dimethylamino)phenylimidazo[1,2-a]pyridines as Radioligands for Imaging β-Amyloid Plaques in Alzheimer’s Disease. J Med Chem 2007; 51:148-58. [DOI: 10.1021/jm700970s] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Jeih-San Liow
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Sami S. Zoghbi
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Jessica Cuevas
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Cesar Baetas
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Jinsoo Hong
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - H. Umesha Shetty
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Nicholas M. Seneca
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Amira K. Brown
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Robert Gladding
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Sebastian S. Temme
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Mary M. Herman
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Robert B. Innis
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Victor W. Pike
- Molecular Imaging Branch and Clinical Brain Disorders Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
143
|
Grant MA, Lazo ND, Lomakin A, Condron MM, Arai H, Yamin G, Rigby AC, Teplow DB. Familial Alzheimer's disease mutations alter the stability of the amyloid beta-protein monomer folding nucleus. Proc Natl Acad Sci U S A 2007; 104:16522-7. [PMID: 17940047 PMCID: PMC2034231 DOI: 10.1073/pnas.0705197104] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Indexed: 12/26/2022] Open
Abstract
Amyloid beta-protein (Abeta) oligomers may be the proximate neurotoxins in Alzheimer's disease (AD). Recently, to elucidate the oligomerization pathway, we studied Abeta monomer folding and identified a decapeptide segment of Abeta, (21)Ala-(22)Glu-(23)Asp-(24)Val-(25)Gly-(26)Ser-(27)Asn-(28)Lys-(29)Gly-(30)Ala, within which turn formation appears to nucleate monomer folding. The turn is stabilized by hydrophobic interactions between Val-24 and Lys-28 and by long-range electrostatic interactions between Lys-28 and either Glu-22 or Asp-23. We hypothesized that turn destabilization might explain the effects of amino acid substitutions at Glu-22 and Asp-23 that cause familial forms of AD and cerebral amyloid angiopathy. To test this hypothesis, limited proteolysis, mass spectrometry, and solution-state NMR spectroscopy were used here to determine and compare the structure and stability of the Abeta(21-30) turn within wild-type Abeta and seven clinically relevant homologues. In addition, we determined the relative differences in folding free energies (DeltaDeltaG(f)) among the mutant peptides. We observed that all of the disease-associated amino acid substitutions at Glu-22 or Asp-23 destabilized the turn and that the magnitude of the destabilization correlated with oligomerization propensity. The Ala21Gly (Flemish) substitution, outside the turn proper (Glu-22-Lys-28), displayed a stability similar to that of the wild-type peptide. The implications of these findings for understanding Abeta monomer folding and disease causation are discussed.
Collapse
Affiliation(s)
- Marianne A. Grant
- *Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Noel D. Lazo
- Gustaf A. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main Street, Worcester, MA 01610
| | - Aleksey Lomakin
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139; and
| | | | - Hiromi Arai
- Department of Neurology, David Geffen School of Medicine, and
| | - Ghiam Yamin
- Department of Neurology, David Geffen School of Medicine, and
| | - Alan C. Rigby
- *Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - David B. Teplow
- Department of Neurology, David Geffen School of Medicine, and
- Molecular Biology Institute and Brain Research Institute, University of California, Los Angeles, CA 90095
| |
Collapse
|
144
|
Wu J, Khan GM, Nichols RA. Dopamine release in prefrontal cortex in response to beta-amyloid activation of alpha7 * nicotinic receptors. Brain Res 2007; 1182:82-9. [PMID: 17935702 DOI: 10.1016/j.brainres.2007.08.079] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2007] [Revised: 08/23/2007] [Accepted: 08/30/2007] [Indexed: 10/22/2022]
Abstract
The levels of soluble beta-amyloid (Abeta) are correlated with symptom severity in Alzheimer's disease. Soluble Abeta has been shown to disrupt synaptic function and it has been proposed that accumulation of soluble Abeta triggers synapse loss over the course of the disease. Numerous studies indicate that soluble Abeta has multiple targets, one of which appears to be the nicotinic acetylcholine receptor, particularly for Abeta concentrations of pM to nM. Moreover, pM to nM soluble Abeta was found to increase presynaptic Ca(2+) levels, suggesting that it may have an impact on neurotransmitter release. In the present study, soluble Abeta was perfused into mouse prefrontal cortex and the effect on the release of dopamine outflow via microdialysis was assessed. In the presence of tetrodotoxin, Abeta(1-42) at 100 nM evoked the release of dopamine to approximately 170% of basal levels. The Abeta(1-42)-evoked dopamine release was sensitive to antagonists of alpha7 nicotinic receptors and was absent in mice harboring a null mutation for the alpha7 nicotinic subunit, but was intact in mice harboring a null mutation for the beta2 nicotinic subunit. The control peptide Abeta(40-1) was without effect. In contrast, Abeta(1-42) at 1-10 pM caused a profound but slowly developing decrease in dopamine outflow. These results suggest that Abeta alters dopamine release in mouse prefrontal cortex, perhaps involving distinct targets as it accumulates during Alzheimer's disease and leading to disruption of synaptic signaling.
Collapse
Affiliation(s)
- Jianlin Wu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102, USA
| | | | | |
Collapse
|
145
|
Arriagada C, Astorga C, Atwater I, Rojas E, Mears D, Caviedes R, Caviedes P. Endosomal abnormalities related to amyloid precursor protein in cholesterol treated cerebral cortex neuronal cells derived from trisomy 16 mice, an animal model of Down syndrome. Neurosci Lett 2007; 423:172-7. [PMID: 17706358 DOI: 10.1016/j.neulet.2007.06.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 06/11/2007] [Accepted: 06/22/2007] [Indexed: 11/22/2022]
Abstract
The CNh and CTb cell lines are derived from the cerebral cortex of normal and trisomy 16 mice, an animal model of human trisomy 21, Down syndrome (DS), and represent in vitro models to study cellular events associated with the human condition. Amyloid precursor protein (APP) plays an important role in the development of neuropathology associated with DS and cholesterol in the amyloidogenic processing of APP. There is also increasing evidence of alterations in the recycling pathway of the early endosome compartment in nervous tissue from DS. In the present study, we report endosomal abnormalities related to amyloid precursor protein in cholesterol-treated CTb cells. Colocalization studies revealed the presence of APP-derived products in early endosomal compartments in both cell lines. Using internalization and immunoprecipitation techniques, differential effects were observed between the normal and trisomic cell lines when treated with cholesterol. Internalization experiments showed that the CTb cell line accumulates internalized APP in intracellular compartments for longer periods of time when compared to the CNh cell line. Immunoprecipitation revealed a differential interaction between the trafficking-related protein Rab4 and APP in the neuronal cell lines CNh and CTb. The present study suggests a putative mechanism by which overexpressed APP accumulates in intracellular compartments related to the endosomal trafficking pathway in individuals with DS, and highlights the usefulness of the CTb cell line as a model to study altered APP metabolism related to this genetic condition.
Collapse
Affiliation(s)
- Christian Arriagada
- ICBM, Program of Anatomy and Developmental Biology, Faculty of Medicine, University of Chile, Casilla 70079, Correo 7, Santiago, Chile.
| | | | | | | | | | | | | |
Collapse
|
146
|
Grimm MOW, Grimm HS, Hartmann T. Amyloid beta as a regulator of lipid homeostasis. Trends Mol Med 2007; 13:337-44. [PMID: 17644432 DOI: 10.1016/j.molmed.2007.06.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 05/25/2007] [Accepted: 06/28/2007] [Indexed: 11/18/2022]
Abstract
The beta-amyloid peptide (A beta) is widely considered to be the molecule that causes Alzheimer's disease (AD). Besides this pathological function of A beta, recently published data reveal that A beta also has an essential physiological role in lipid homeostasis. Cholesterol increases A beta production, and conversely A beta production causes a decrease in cholesterol synthesis. The latter appears to be mediated by the inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR), a key enzyme in cholesterol synthesis, in an action similar to that of statins. Moreover, A beta regulates sphingolipid metabolism by directly activating sphingomyelinases (SMases). This review summarizes the molecular basis for the known physiological functions of A beta and amyloid precursor protein (APP), the roles of A beta and APP in lipid homeostasis and the medical implications of addressing lipid homeostasis in respect to AD. This knowledge might provide new insights for current and future therapeutic approaches to AD.
Collapse
Affiliation(s)
- Marcus O W Grimm
- Universität des Saarlandes, Kirrberger Str. 61.4, D-66421 Homburg, Germany
| | | | | |
Collapse
|
147
|
Boddaert J, Kinugawa K, Lambert JC, Boukhtouche F, Zoll J, Merval R, Blanc-Brude O, Mann D, Berr C, Vilar J, Garabedian B, Journiac N, Charue D, Silvestre JS, Duyckaerts C, Amouyel P, Mariani J, Tedgui A, Mallat Z. Evidence of a role for lactadherin in Alzheimer's disease. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:921-9. [PMID: 17322377 PMCID: PMC1864868 DOI: 10.2353/ajpath.2007.060664] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lactadherin is a secreted extracellular matrix protein expressed in phagocytes and contributes to the removal of apoptotic cells. We examined lactadherin expression in brain sections of patients with or without Alzheimer's disease and studied its role in the phagocytosis of amyloid beta-peptide (Abeta). Cells involved in Alzheimer's disease, including vascular smooth muscle cells, astrocytes, and microglia, showed a time-related increase in lactadherin production in culture. Quantitative analysis of the level of lactadherin showed a 35% reduction in lactadherin mRNA expression in the brains of patients with Alzheimer's disease (n = 52) compared with age-matched controls (n = 58; P = 0.003). Interestingly, lactadherin protein was detected in the brains of patients with Alzheimer's disease and controls, with low expression in areas rich in senile plaques and marked expression in areas without Abeta deposition. Using surface plasmon resonance, we observed a direct protein-protein interaction between recombinant lactadherin and Abeta 1-42 peptide in vitro. Lactadherin deficiency or its neutralization using specific antibodies significantly prevented Abeta 1-42 phagocytosis by murine and human macrophages. In conclusion, lactadherin plays an important role in the phagocytosis of Abeta 1-42 peptide, and its expression is reduced in Alzheimer's disease. Alterations in lactadherin production/function may contribute to the initiation and/or progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Jacques Boddaert
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche Cardiovasculaire Lariboisière, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Szymańska I, Radecka H, Radecki J, Kaliszan R. Electrochemical impedance spectroscopy for study of amyloid β-peptide interactions with (−) nicotine ditartrate and (−) cotinine. Biosens Bioelectron 2007; 22:1955-60. [PMID: 17000100 DOI: 10.1016/j.bios.2006.08.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 07/27/2006] [Accepted: 08/18/2006] [Indexed: 11/24/2022]
Abstract
Immobilization of amyloid beta (Abeta) (1-40) peptide on Au-colloid modified gold electrodes has been studied. Colloidal Au was self-assembled onto gold electrodes through the thiol groups of 1,6-hexanedithiol monolayer. Next, buffered aqueous solution of Abeta (1-40) peptide existing in the beta-sheet structure in the acidic media was dropped on the electrode surface. Each step of electrode modification has been confirmed with cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS). The changes of the resistance of the layer with deposited Abeta (1-40) peptide, occurred under stimulation by different concentration of (-) nicotine ditartrate and (-) cotinine were measured with EIS and were used for the calculation of association constants. The gentle measuring conditions applied in electrochemical impedance spectroscopy, together with suitable environment for biomolecules immobilization created by Au-colloid, might be recommended as the analytical tool for assessing the effectiveness of potential drugs used in Alzheimer's disease (AD) therapy.
Collapse
Affiliation(s)
- Iwona Szymańska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Division of Food Sciences, Tuwima 10, 10-747 Olsztyn, Poland
| | | | | | | |
Collapse
|
149
|
Abstract
Proteolytic processing of the amyloid precursor protein (APP) is modulated by the action of enzymes alpha-, beta- and gamma-secretases, with the latter two mediating the amyloidogenic production of amyloid-beta (Abeta). Cellular modulators of APP processing are well known from studies of genetic mutations (such as those found in APP and presenilins) or polymorphisms (such as the apolipoprotein E4 epsilon-allele) that predisposes an individual to early or late-onset Alzheimer's disease. In recent years, several classes of molecule with modulating functions in APP processing and Abeta secretion have emerged. These include the neuronal Munc-18 interacting proteins (Mints)/X11s, members of the reticulon family (RTN-3 and RTN-4/Nogo-B), the Nogo-66 receptor (NgR), the peptidyl-prolyl isomerase Pin1 and the Rho family GTPases and their effectors. Mints and NgR bind to APP directly, while RTN3 and Nogo-B interact with the beta-secretase BACE1. Phosphorylated APP is a Pin1 substrate, which binds to its phosphor-Thr668-Pro motif. These interactions by and large resulted in a reduction of Abeta generation both in vitro and in vivo. Inhibition of Rho and Rho-kinase (ROCK) activity may underlie the ability of non-steroidal anti-inflammatory drugs and statins to reduce Abeta production, a feat which could also be achieved by Rac1 inhibition. Detailed understanding of the underlying mechanisms of action of these novel modulators of APP processing, as well as insights into the molecular neurological basis of how Abeta impairs leaning and memory, will open up multiple avenues for the therapeutic intervention of Alzheimer's disease.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | |
Collapse
|
150
|
Miyata S, Nishimura Y, Nakashima T. Perineuronal nets protect against amyloid beta-protein neurotoxicity in cultured cortical neurons. Brain Res 2007; 1150:200-6. [PMID: 17397805 DOI: 10.1016/j.brainres.2007.02.066] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 02/26/2007] [Accepted: 02/27/2007] [Indexed: 11/24/2022]
Abstract
Perineuronal nets (PNs) consisting of chondroitin sulfate proteoglycans (CSPGs) and hyaluronic acid are associated with distinct neuronal populations in mammalian brain. Cortical areas abundant in PNs have been known to be less affected by neurotoxicity in human Alzheimer's disease. In the present study, we examined whether PNs protect the neurotoxicity caused by amyloid beta-protein (Abeta), a major constituent of senile plaques in Alzheimer's disease using cortical neurons of dissociated culture. Double labeling experiments using confocal microscopy showed that the neurons associated with PNs were visualized with the anti-CSPG antibody in dissociated cortical culture. The analysis of reverse transcription-polymerase chain reaction revealed that mRNA expression of chondroitin sulfotransferases, CSPG-specific enzymes, was detected in neuronal culture, indicating that cultured cortical neurons are able to synthesize CSPGs and construct PNs structure. The treatment of Abeta1-42 showed significant neurotoxicity on PNs-free cortical neurons, however, it did not reveal neurotoxicity on PNs-associated neurons. Moreover, it was shown that the treatment of Abeta1-42 was able to kill PNs-associated neurons after the removal of chondroitin sulfate (CS) glycosaminoglycans with chondroitinase ABC. The treatment of glutamate killed not only PNs-free cortical neurons but also PNs-associated neurons. These results suggest that CS glycosaminoglycans on PNs are responsible for protecting neurons from Abeta1-42 neurotoxicity.
Collapse
Affiliation(s)
- Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| | | | | |
Collapse
|