501
|
O’Connor T, Doherty-Sadleir KR, Maus E, Velliquette RA, Zhao J, Cole SL, Eimer WA, Hitt B, Bembinster LA, Lammich S, Lichtenthaler SF, Hébert SS, De Strooper B, Haass C, Bennett DA, Vassar R. Phosphorylation of the translation initiation factor eIF2alpha increases BACE1 levels and promotes amyloidogenesis. Neuron 2008; 60:988-1009. [PMID: 19109907 PMCID: PMC2667382 DOI: 10.1016/j.neuron.2008.10.047] [Citation(s) in RCA: 336] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 09/16/2008] [Accepted: 10/17/2008] [Indexed: 10/21/2022]
Abstract
beta-site APP cleaving enzyme-1 (BACE1), the rate-limiting enzyme for beta-amyloid (Abeta) production, is elevated in Alzheimer's disease (AD). Here, we show that energy deprivation induces phosphorylation of the translation initiation factor eIF2alpha (eIF2alpha-P), which increases the translation of BACE1. Salubrinal, an inhibitor of eIF2alpha-P phosphatase PP1c, directly increases BACE1 and elevates Abeta production in primary neurons. Preventing eIF2alpha phosphorylation by transfection with constitutively active PP1c regulatory subunit, dominant-negative eIF2alpha kinase PERK, or PERK inhibitor P58(IPK) blocks the energy-deprivation-induced BACE1 increase. Furthermore, chronic treatment of aged Tg2576 mice with energy inhibitors increases levels of eIF2alpha-P, BACE1, Abeta, and amyloid plaques. Importantly, eIF2alpha-P and BACE1 are elevated in aggressive plaque-forming 5XFAD transgenic mice, and BACE1, eIF2alpha-P, and amyloid load are correlated in humans with AD. These results strongly suggest that eIF2alpha phosphorylation increases BACE1 levels and causes Abeta overproduction, which could be an early, initiating molecular mechanism in sporadic AD.
Collapse
Affiliation(s)
- Tracy O’Connor
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Erika Maus
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rodney A. Velliquette
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jie Zhao
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sarah L. Cole
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - William A. Eimer
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Brian Hitt
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Leslie A. Bembinster
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sven Lammich
- Center for Integrated Protein Science Munich and Adolf-Butenandt-Institute, Department of Biochemistry, Laboratory for Neurodegenerative Disease Research, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan F. Lichtenthaler
- Center for Integrated Protein Science Munich and Adolf-Butenandt-Institute, Department of Biochemistry, Laboratory for Neurodegenerative Disease Research, Ludwig-Maximilians-University, Munich, Germany
| | - Sébastien S. Hébert
- Center for Human Genetics, Katholieke Universiteit Leuven and Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Bart De Strooper
- Center for Human Genetics, Katholieke Universiteit Leuven and Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Christian Haass
- Center for Integrated Protein Science Munich and Adolf-Butenandt-Institute, Department of Biochemistry, Laboratory for Neurodegenerative Disease Research, Ludwig-Maximilians-University, Munich, Germany
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL
| | - Robert Vassar
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
502
|
Yu Y, Jia J. Lack of association between the polymorphisms of beta-site APP-cleaving enzyme 2 (BACE2) 5'-flanking region and sporadic Alzheimer's disease. Brain Res 2008; 1257:10-5. [PMID: 19124009 DOI: 10.1016/j.brainres.2008.12.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 11/18/2008] [Accepted: 12/15/2008] [Indexed: 10/21/2022]
Abstract
Amyloid beta-peptide (A beta) plays a central role in the pathogenesis of Alzheimer's disease (AD). A beta is produced by sequential cleavage of the amyloid precursor protein (APP) by two enzymes referred to as beta- and gamma-secretase. beta-secretase is of more importance, as it catalyses the rate-limiting step in the production of A beta. Although beta-site APP-cleaving enzyme 1 (BACE1) is known to cleave APP at the beta-secretase site as required for the generation of A beta, the role of its homologue BACE2 is controversial. For seeking the correlation of the BACE2 promoter with sporadic AD (SAD), we performed a case-control study in a Chinese Han population. In the study, we sequenced the 2641 bp fragment of the 5'-flanking region of BACE2 gene and found three polymorphisms which are -320C/- (rs11316732), -1541A/T (rs9975138) and -1904C/T (rs28656880). Definitive genotyping these markers and apolipoprotein E (APOE) polymorphism were surveyed using restriction enzyme digestion and direct sequencing in 359 SAD patients and 334 controls. We failed to find any association between these three polymorphisms and SAD even after statistical adjustment for age, gender and APOE epsilon 4 status. Our data do not support that there is a linkage between the 5'-flanking region polymorphisms of BACE2 and SAD in the Chinese Han population.
Collapse
Affiliation(s)
- Yueyi Yu
- Department of Neurology, Xuan Wu Hospital of the Capital Medical University, Key Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, PR China
| | | |
Collapse
|
503
|
Vetrivel KS, Meckler X, Chen Y, Nguyen PD, Seidah NG, Vassar R, Wong PC, Fukata M, Kounnas MZ, Thinakaran G. Alzheimer disease Abeta production in the absence of S-palmitoylation-dependent targeting of BACE1 to lipid rafts. J Biol Chem 2008; 284:3793-803. [PMID: 19074428 DOI: 10.1074/jbc.m808920200] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Alzheimer disease beta-amyloid (Abeta) peptides are generated via sequential proteolysis of amyloid precursor protein (APP) by BACE1 and gamma-secretase. A subset of BACE1 localizes to cholesterol-rich membrane microdomains, termed lipid rafts. BACE1 processing in raft microdomains of cultured cells and neurons was characterized in previous studies by disrupting the integrity of lipid rafts by cholesterol depletion. These studies found either inhibition or elevation of Abeta production depending on the extent of cholesterol depletion, generating controversy. The intricate interplay between cholesterol levels, APP trafficking, and BACE1 processing is not clearly understood because cholesterol depletion has pleiotropic effects on Golgi morphology, vesicular trafficking, and membrane bulk fluidity. In this study, we used an alternate strategy to explore the function of BACE1 in membrane microdomains without altering the cellular cholesterol level. We demonstrate that BACE1 undergoes S-palmitoylation at four Cys residues at the junction of transmembrane and cytosolic domains, and Ala substitution at these four residues is sufficient to displace BACE1 from lipid rafts. Analysis of wild type and mutant BACE1 expressed in BACE1 null fibroblasts and neuroblastoma cells revealed that S-palmitoylation neither contributes to protein stability nor subcellular localization of BACE1. Surprisingly, non-raft localization of palmitoylation-deficient BACE1 did not have discernible influence on BACE1 processing of APP or secretion of Abeta. These results indicate that post-translational S-palmitoylation of BACE1 is not required for APP processing, and that BACE1 can efficiently cleave APP in both raft and non-raft microdomains.
Collapse
Affiliation(s)
- Kulandaivelu S Vetrivel
- Department of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
504
|
Abstract
The amyloid-beta peptide (Abeta), implicated in the pathogenesis of Alzheimer's disease (AD), is produced through sequential proteolysis of the Abeta precursor protein (APP) by beta- and gamma-secretases. Thus, blocking either of these two proteases, directly or indirectly, is potentially worthwhile toward developing AD therapeutics. beta-Secretase is a membrane-tethered pepsin-like aspartyl protease suitable for structure-based design, whereas gamma-secretase is an unusual, heterotetrameric membrane-embedded aspartyl protease. While gamma-secretase inhibitors entered clinical trials first due to their superior pharmacological properties (for example, brain penetration) over beta-secretase inhibitors, it has since become clear that gamma-secretase inhibitors can cause mechanism-based toxicities owing to interference with the proteolysis of another gamma-secretase substrate, the Notch receptor. Strategies for targeting Abeta production at the gamma-secretase level without blocking Notch signalling will be discussed. Other strategies utilizing cell-based screening have led to the identification of novel Abeta lowering agents that likewise leave Notch proteolysis intact. The mechanism by which these agents lower Abeta is unknown, but these compounds may ultimately reveal new targets for AD therapeutics.
Collapse
Affiliation(s)
- Michael S Wolfe
- Center for Neurologic Diseases, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
505
|
Böhme L, Hoffmann T, Manhart S, Wolf R, Demuth HU. Isoaspartate-containing amyloid precursor protein-derived peptides alter efficacy and specificity of potential beta-secretases. Biol Chem 2008; 389:1055-66. [PMID: 18979630 DOI: 10.1515/bc.2008.125] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neuritic plaques of Alzheimer patients are composed of multiple protein components. Among them, the amyloid beta-peptides (Abeta) 1-40/42 and further N- and C-terminally modified fragments of Abeta are highly abundant. Most prominent are the isoaspartate (isoAsp)-Abeta peptides and pyroglutamyl (pGlu)-Abeta. While pGlu-Abeta can only be formed from an N-terminal glutamate by glutaminyl cyclase, spontaneous isoAsp-isomerization cannot occur at an N-terminal aspartate of peptides. This means that isoAsp-Abeta formation must precede proteolysis of the amyloid precursor protein (APP). Abeta generation from APP by beta- and gamma-secretases initiates the amyloid peptide aggregation and deposition process. Two aspartate proteases have been identified as secretases: BACE-1 (beta-site amyloid precursor protein cleaving enzyme) and the intramembrane gamma-secretase multiprotein complex. However, recent evidence supports more than one beta-secretase initiating this cascade. Formation of Abeta1-40/42 was predominantly studied by expression of mutated human APP sequences in cell culture and transgenic animals, generating Abeta fragments that did not contain such multiple posttranslational modifications as in Alzheimer's disease. This prompted us to investigate the catalytic turnover of Asp- or isoAsp-containing APP-derived peptide sequences by BACE-1 and cathepsin B, another potential beta-secretase. While cathepsin B is more effective than BACE-1 in processing the Asp-containing peptide derivatives, only cathepsin B can cleave the isoAsp-containing peptides, which occurs with high catalytic efficiency.
Collapse
Affiliation(s)
- Livia Böhme
- Probiodrug AG, Biocenter, Weinbergweg 22, D-06120 Halle/Saale, Germany
| | | | | | | | | |
Collapse
|
506
|
Bäck M, Nyhlén J, Kvarnström I, Appelgren S, Borkakoti N, Jansson K, Lindberg J, Nyström S, Hallberg A, Rosenquist Å, Samuelsson B. Design, synthesis and SAR of potent statine-based BACE-1 inhibitors: Exploration of P1 phenoxy and benzyloxy residues. Bioorg Med Chem 2008; 16:9471-86. [DOI: 10.1016/j.bmc.2008.09.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 09/10/2008] [Accepted: 09/16/2008] [Indexed: 10/21/2022]
|
507
|
Qing H, He G, Ly PTT, Fox CJ, Staufenbiel M, Cai F, Zhang Z, Wei S, Sun X, Chen CH, Zhou W, Wang K, Song W. Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer's disease mouse models. ACTA ACUST UNITED AC 2008; 205:2781-9. [PMID: 18955571 PMCID: PMC2585842 DOI: 10.1084/jem.20081588] [Citation(s) in RCA: 281] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Neuritic plaques in the brains are one of the pathological hallmarks of Alzheimer's disease (AD). Amyloid β-protein (Aβ), the central component of neuritic plaques, is derived from β-amyloid precursor protein (APP) after β- and γ-secretase cleavage. The molecular mechanism underlying the pathogenesis of AD is not yet well defined, and there has been no effective treatment for AD. Valproic acid (VPA) is one of the most widely used anticonvulsant and mood-stabilizing agents for treating epilepsy and bipolar disorder. We found that VPA decreased Aβ production by inhibiting GSK-3β–mediated γ-secretase cleavage of APP both in vitro and in vivo. VPA treatment significantly reduced neuritic plaque formation and improved memory deficits in transgenic AD model mice. We also found that early application of VPA was important for alleviating memory deficits of AD model mice. Our study suggests that VPA may be beneficial in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Hong Qing
- Department of Psychiatry, Townsend Family Laboratories, Brain Research Center, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
508
|
Varvel NH, Bhaskar K, Patil AR, Pimplikar SW, Herrup K, Lamb BT. Abeta oligomers induce neuronal cell cycle events in Alzheimer's disease. J Neurosci 2008; 28:10786-93. [PMID: 18945886 PMCID: PMC2680286 DOI: 10.1523/jneurosci.2441-08.2008] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 08/04/2008] [Accepted: 09/06/2008] [Indexed: 01/07/2023] Open
Abstract
Neurons subject to degeneration in Alzheimer's disease (AD) exhibit evidence of re-entry into a mitotic cell cycle even before the development of substantial AD brain pathology. In efforts to identify the initiating factors underlying these cell cycle events (CCEs), we have characterized the appearance of the neuronal CCEs in the genomic-based R1.40 transgenic mouse model of AD. Notably, R1.40 mice exhibit neuronal CCEs in a reproducible temporal and spatial pattern that recapitulates the neuronal vulnerability seen in human AD. Neuronal CCEs first appear at 6 months in the frontal cortex layers II/III. This is 6-8 months before detectable amyloid beta (Abeta) deposition, suggesting that specific amyloid precursor protein (APP) processing products are responsible for the induction of neuronal CCEs. Furthermore, a reduction in the levels of Abeta (achieved by shifting the genetic background from C57BL/6 to the DBA/2 mouse strain) dramatically delays the appearance of neuronal CCEs. More significantly, elimination of beta-secretase activity blocks the appearance of CCEs, providing direct genetic evidence that the amyloidogenic processing of APP is required for the induction of CCEs. Finally, in vitro preparations of oligomeric, but not monomeric, Abeta induce DNA synthesis in dissociated cortical neurons, and this response is blocked by antioligomer specific antibodies. Together, our data suggest that low molecular weight aggregates of Abeta induce neuronal cell cycle re-entry in mouse models of Alzheimer's disease.
Collapse
Affiliation(s)
- Nicholas H. Varvel
- Department of Neurosciences, NC30, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195-0001
- Departments of Neurosciences and
| | - Kiran Bhaskar
- Department of Neurosciences, NC30, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195-0001
| | - Anita R. Patil
- Department of Neurosciences, NC30, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195-0001
| | - Sanjay W. Pimplikar
- Department of Neurosciences, NC30, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195-0001
| | - Karl Herrup
- Department of Cell Biology and Neuroscience, Nelson Biological Laboratories, Rutgers, The State University of New Jersey, Busch Campus, Piscataway, New Jersey 08854-8082
| | - Bruce T. Lamb
- Department of Neurosciences, NC30, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195-0001
- Departments of Neurosciences and
- Genetics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-4955, and
| |
Collapse
|
509
|
Woodard-Grice AV, McBrayer AC, Wakefield JK, Zhuo Y, Bellis SL. Proteolytic shedding of ST6Gal-I by BACE1 regulates the glycosylation and function of alpha4beta1 integrins. J Biol Chem 2008; 283:26364-73. [PMID: 18650447 PMCID: PMC2546544 DOI: 10.1074/jbc.m800836200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 07/21/2008] [Indexed: 12/11/2022] Open
Abstract
Differentiation of monocytes into macrophages is accompanied by increased cell adhesiveness, due in part to the activation of alpha4beta1 integrins. Here we report that the sustained alpha4beta1 activation associated with macrophage differentiation results from expression of beta1 integrin subunits that lack alpha2-6-linked sialic acids, a carbohydrate modification added by the ST6Gal-I sialyltransferase. During differentiation of U937 monocytic cells and primary human CD14(+) monocytes, ST6Gal-I is down-regulated, leading to beta1 hyposialylation and enhanced alpha4beta1-dependent VCAM-1 binding. Importantly, ST6Gal-I down-regulation results from cleavage by the BACE1 secretase, which we show is dramatically up-regulated during macrophage differentiation. BACE1 up-regulation, ST6Gal-I shedding, beta1 hyposialylation, and alpha4beta1-dependent VCAM-1 binding are all temporally correlated and share the same signaling mechanism (protein kinase C/Ras/ERK). Preventing ST6Gal-I down-regulation (and therefore integrin hyposialylation), through BACE1 inhibition or ST6Gal-I constitutive overexpression, eliminates VCAM-1 binding. Similarly, preventing integrin hyposialylation inhibits a differentiation-induced increase in the expression of an activation-dependent conformational epitope on the beta1 subunit. Collectively, these results describe a novel mechanism for alpha4beta1 regulation and further suggest an unanticipated role for BACE1 in macrophage function.
Collapse
Affiliation(s)
- Alencia V. Woodard-Grice
- Department of Physiology and Biophysics
and Department of Surgery, University of Alabama
at Birmingham, Birmingham, Alabama 35294 and
Tranzyme Pharma, Birmingham, Alabama 35294
| | - Alexis C. McBrayer
- Department of Physiology and Biophysics
and Department of Surgery, University of Alabama
at Birmingham, Birmingham, Alabama 35294 and
Tranzyme Pharma, Birmingham, Alabama 35294
| | - John K. Wakefield
- Department of Physiology and Biophysics
and Department of Surgery, University of Alabama
at Birmingham, Birmingham, Alabama 35294 and
Tranzyme Pharma, Birmingham, Alabama 35294
| | - Ya Zhuo
- Department of Physiology and Biophysics
and Department of Surgery, University of Alabama
at Birmingham, Birmingham, Alabama 35294 and
Tranzyme Pharma, Birmingham, Alabama 35294
| | - Susan L. Bellis
- Department of Physiology and Biophysics
and Department of Surgery, University of Alabama
at Birmingham, Birmingham, Alabama 35294 and
Tranzyme Pharma, Birmingham, Alabama 35294
| |
Collapse
|
510
|
Iben LG, Kopcho L, Marcinkeviciene J, Zheng C, Thompson LA, Albright CF, Toyn JH. [3H]BMS-599240 — A novel tritiated ligand for the characterization of BACE1 inhibitors. Eur J Pharmacol 2008; 593:10-5. [DOI: 10.1016/j.ejphar.2008.06.112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Revised: 06/19/2008] [Accepted: 06/27/2008] [Indexed: 12/20/2022]
|
511
|
Wang H, Song L, Laird F, Wong PC, Lee HK. BACE1 knock-outs display deficits in activity-dependent potentiation of synaptic transmission at mossy fiber to CA3 synapses in the hippocampus. J Neurosci 2008; 28:8677-81. [PMID: 18753368 PMCID: PMC2728626 DOI: 10.1523/jneurosci.2440-08.2008] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 07/11/2008] [Accepted: 07/19/2008] [Indexed: 11/21/2022] Open
Abstract
beta-Amyloid precursor protein cleavage enzyme 1 (BACE1) has been identified as a major neuronal beta-secretase critical for the formation of beta-amyloid (Abeta) peptide, which is thought responsible for the pathology of Alzheimer's disease (AD). Therefore, BACE1 is one of the key therapeutic targets that can prevent the progression of AD. Previous studies showed that knocking out the BACE1 gene prevents Abeta formation, but results in behavioral deficits and specific synaptic dysfunctions at Schaffer collateral to CA1 synapses. However, BACE1 protein is most highly expressed at the mossy fiber projections in CA3. Here, we report that BACE1 knock-out mice display reduced presynaptic function, as measured by an increase in paired-pulse facilitation ratio. More dramatically, mossy fiber long-term potentiation (LTP), which is normally expressed via an increase in presynaptic release, was eliminated in the knock-outs. Although long-term depression was slightly larger in the BACE1 knock-outs, it could not be reversed. The specific deficit in mossy fiber LTP was upstream of cAMP signaling and could be "rescued" by transiently elevating extracellular Ca2+ concentration. These results suggest that BACE1 may play a critical role in regulating presynaptic function, especially activity-dependent strengthening of presynaptic release, at mossy fiber synapses.
Collapse
Affiliation(s)
- Hui Wang
- Department of Biology, College of Chemical and Life Sciences, and
- Neuroscience and Cognitive Science Program, University of Maryland, College Park, Maryland 20742, and
| | - Lihua Song
- Department of Biology, College of Chemical and Life Sciences, and
| | - Fiona Laird
- Departments of Pathology and
- Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Philip C. Wong
- Departments of Pathology and
- Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Hey-Kyoung Lee
- Department of Biology, College of Chemical and Life Sciences, and
- Neuroscience and Cognitive Science Program, University of Maryland, College Park, Maryland 20742, and
| |
Collapse
|
512
|
Limongelli V, Marinelli L, Cosconati S, Braun HA, Schmidt B, Novellino E. Ensemble-docking approach on BACE-1: pharmacophore perception and guidelines for drug design. ChemMedChem 2008; 2:667-78. [PMID: 17407105 DOI: 10.1002/cmdc.200600314] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Beta-secretase (BACE-1), a key enzyme in the etiopathogenesis and progression of Alzheimer disease, is the focus of medicinal chemistry efforts both in the pharmaceutical industry and in academia. Despite the availability of diverse peptidomimetic BACE-1 inhibitors, nonpeptidic compounds suitable for oral delivery and transport across the blood brain barrier are in great demand. Herein, a number of active and structurally diverse inhibitors were selected and subjected to an ensemble-docking process into five BACE-1 X-ray structures. The calculated bioactive conformations of these inhibitors allowed us to build an exhaustive pharmacophore model, which captures both the common geometric and electronic features essential for enzyme inhibition. The model is intended to aid the rational design of new BACE-1 inhibitors. Furthermore, a comparison of BACE/cathepsin D X-ray structures was made to provide guidelines for the design of BACE-selective inhibitors.
Collapse
Affiliation(s)
- Vittorio Limongelli
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università di Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | | | | | | | | | | |
Collapse
|
513
|
Böhme L, Hoffmann T, Manhart S, Wolf R, Demuth HU. Isoaspartate containing amyloid precursor protein derived peptides alter efficacy and specificity of potential β-secretases. Biol Chem 2008. [DOI: 10.1515/bc.2008.125_bchm.just-accepted] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
514
|
Hook V, Schechter I, Demuth HU, Hook G. Alternative pathways for production of β-amyloid peptides of Alzheimer's disease. Biol Chem 2008. [DOI: 10.1515/bc.2008.124_bchm.just-accepted] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
515
|
Husain MM, Trevino K, Siddique H, McClintock SM. Present and prospective clinical therapeutic regimens for Alzheimer's disease. Neuropsychiatr Dis Treat 2008; 4:765-77. [PMID: 19043521 PMCID: PMC2536544 DOI: 10.2147/ndt.s2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder that produces cognitive impairments that increase in severity as the disease progresses. The clinical symptoms are related to the presence of neuritic plaques and neurofibrillary tangles in the cerebral cortex which represent the pathophysiological hallmarks of AD. The debilitating nature of the disease can result in clinical burden for the patient, emotional strain for those that care for patients with Alzheimer's, and significant financial burden to society. The goals of current treatments, such as cholinesterase inhibitors and N-methyl-D-aspartate receptor antagonist, are to reduce the severity or slow the progression of cognitive symptoms. Although these treatments have demonstrated modest clinical benefit, they are unable to prevent, prohibit, or reverse the underlying pathophysiology of AD. Considerable progress has been made toward the development of disease-modifying treatments. Treatments currently under development mainly target the production, aggregation, and removal of existing amyloid beta-peptide aggregates which are believed to instigate the overall development of the neuropathology. Additional strategies that target tau pathology are being studied to promote neural protection against AD pathology. The current research has continued to expand our knowledge toward the development of disease modifying Alzheimer's therapies; however, no specific treatment strategy capable of demonstrating empirical efficacy and safety has yet to emerge.
Collapse
Affiliation(s)
- Mustafa M Husain
- Department of Psychiatry, University of Texas Southwestern Medical Center Dallas, TX, USA
| | | | | | | |
Collapse
|
516
|
Wu G, Sankaranarayanan S, Tugusheva K, Kahana J, Seabrook G, Shi XP, King E, Devanarayan V, Cook JJ, Simon AJ. Decrease in age-adjusted cerebrospinal fluid β-secretase activity in Alzheimer's subjects. Clin Biochem 2008; 41:986-96. [DOI: 10.1016/j.clinbiochem.2008.04.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 03/12/2008] [Accepted: 04/20/2008] [Indexed: 12/30/2022]
|
517
|
Hook V, Schechter I, Demuth HU, Hook G. Alternative pathways for production of beta-amyloid peptides of Alzheimer's disease. Biol Chem 2008; 389:993-1006. [PMID: 18979625 PMCID: PMC2654319 DOI: 10.1515/bc.2008.124] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This highlight article describes three Alzheimer's disease (AD) studies presented at the 5th General Meeting of the International Proteolysis Society that address enzymatic mechanisms for producing neurotoxic beta-amyloid (Abeta) peptides. One group described the poor kinetics of BACE 1 for cleaving the wild-type (WT) beta-secretase site of APP found in most AD patients. They showed that cathepsin D displays BACE 1-like specificity and cathepsin D is 280-fold more abundant in human brain than BACE 1. Nevertheless, as BACE 1 and cathepsin D show poor activity towards the WT beta-secretase site, they suggested continuing the search for additional beta-secretase(s). The second group reported cathepsin B as an alternative beta-secretase possessing excellent kinetic efficiency and specificity for the WT beta-secretase site. Significantly, inhibitors of cathepsin B improved memory, with reduced amyloid plaques and decreased Abeta(40/42) in brains of AD animal models expressing amyloid precursor protein containing the WT beta-secretase site. The third group addressed isoaspartate and pyroglutamate (pGlu) posttranslational modifications of Abeta. Results showed that cathepsin B, but not BACE 1, efficiently cleaves the WT beta-secretase isoaspartate site. Furthermore, cyclization of N-terminal Glu by glutaminyl cyclase generates highly amyloidogenic pGluAbeta(3-40/42). These presentations suggest cathepsin B and glutaminyl cyclase as potential new AD therapeutic targets.
Collapse
Affiliation(s)
- Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA.
| | | | | | | |
Collapse
|
518
|
Cole SL, Vassar R. The role of amyloid precursor protein processing by BACE1, the beta-secretase, in Alzheimer disease pathophysiology. J Biol Chem 2008; 283:29621-5. [PMID: 18650431 DOI: 10.1074/jbc.r800015200] [Citation(s) in RCA: 197] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Amyloid plaques, composed of the amyloid beta-protein (Abeta), are hallmark neuropathological lesions in Alzheimer disease (AD) brain. Abeta fulfills a central role in AD pathogenesis, and reduction of Abeta levels should prove beneficial for AD treatment. Abeta generation is initiated by proteolysis of amyloid precursor protein (APP) by the beta-secretase enzyme BACE1. Bace1 knockout (Bace1(-/-)) mice have validated BACE1 as the authentic beta-secretase in vivo. BACE1 is essential for Abeta generation and represents a suitable drug target for AD therapy, especially because this enzyme is up-regulated in AD. However, although initial data indicated that Bace1(-/-) mice lack an overt phenotype, the BACE1-mediated processing of APP and other substrates may be important for specific biological processes. In this minireview, topics range from the initial identification of BACE1 to the fundamental knowledge gaps that remain in our understanding of this protease. We address pertinent questions such as putative causes of BACE1 elevation in AD and discuss why, nine years since the identification of BACE1, treatments that address the underlying pathological mechanisms of AD are still lacking.
Collapse
Affiliation(s)
- Sarah L Cole
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | |
Collapse
|
519
|
BACE1 inhibitory effects of lavandulyl flavanones from Sophora flavescens. Bioorg Med Chem 2008; 16:6669-74. [DOI: 10.1016/j.bmc.2008.05.080] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 05/30/2008] [Accepted: 05/31/2008] [Indexed: 11/27/2022]
|
520
|
Abstract
beta-Secretase (memapsin 2, BACE1) is an attractive target for the development of inhibitor drugs to treat Alzheimer's disease (AD). Not only does this protease function at the first step in the pathway leading to the production of amyloid-beta (Abeta), its gene deletion produces only mild phenotypes. In addition, beta-secretase is an aspartic protease whose mechanism and inhibition are well known. The development of beta-secretase inhibitors, actively pursued over the last seven years, has been slow, due to the difficulty in combining the required properties in a single inhibitor molecule. Steady progress in this field, however, has brought about inhibitors that contain many targeted characteristics. In this review, we describe the strategy of structure-based inhibitor evolution in the development of beta-secretase inhibitor drug. The current status of the field offers grounds for some optimism, in that beta-secretase inhibitors have been shown to reduce brain Abeta and to rescue the cognitive decline in transgenic AD mice, and an orally available beta-secretase inhibitor drug candidate is in clinical trial. With this knowledge base, it seems reasonable to expect that more drug candidates will be tested in human, and then successful disease-modifying drugs may ultimately emerge from this target.
Collapse
Affiliation(s)
- Arun K. Ghosh
- grid.169077.e0000000419372197Departments of Chemistry and Medicinal Chemistry, Purdue University, 47907 West Lafayette, Indiana
| | - Sandra Gemma
- grid.169077.e0000000419372197Departments of Chemistry and Medicinal Chemistry, Purdue University, 47907 West Lafayette, Indiana
| | - Jordan Tang
- grid.266902.90000000121793618Oklahoma Medical Research Foundation, University of Oklahoma Health Science Center, 73104 Oklahoma City, Oklahoma
- grid.266902.90000000121793618Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, 73104 Oklahoma City, Oklahoma
- grid.274264.10000000085276890Protein Studies Research Program, MS 28, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, 73104 Oklahoma City, OK
| |
Collapse
|
521
|
Mowrer KR, Wolfe MS. Promotion of BACE1 mRNA Alternative Splicing Reduces Amyloid β-Peptide Production. J Biol Chem 2008; 283:18694-701. [DOI: 10.1074/jbc.m801322200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
522
|
Barten DM, Albright CF. Therapeutic strategies for Alzheimer's disease. Mol Neurobiol 2008; 37:171-86. [PMID: 18581273 DOI: 10.1007/s12035-008-8031-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Accepted: 05/27/2008] [Indexed: 12/22/2022]
Abstract
Therapeutic approaches for Alzheimer's disease (AD) are guided by four disease characteristics: amyloid plaques, neurofibrillar tangles (NFT), neurodegeneration, and dementia. Amyloid plaques are composed largely of 4 kDa beta-amyloid (Abeta) peptides, with the more amyloidogenic, 42 amino acid form (Abeta42) as the primary species. Because multiple, rare mutations that cause early-onset, familial AD lead to increased production or aggregation of Abeta42, amyloid therapeutics aim to reduce the amount of toxic Abeta42 aggregates. Amyloid-based therapies include gamma-secretase inhibitors and modulators, BACE inhibitors, aggregation blockers, catabolism inducers, and anti-Abeta biologics. Tangles are composed of paired helical filaments of hyperphosphorylated tau protein. Tau-based therapeutics include kinase inhibitors, microtubule stabilizers, and catabolism inducers. Therapeutic strategies for neurodegeneration target multiple mechanisms, including excitotoxicity, mitochondrial dysfunction, oxidative damage, and inflammation or stimulation of neuronal viability. Although not disease modifying, cognition enhancers are important to treat the symptom of dementia. Strategies for cognition enhancement include cholinesterase inhibitors, and other approaches to enhance the signaling of cholinergic and glutamatergic neurons. In summary, plaques, tangles, neurodegeneration and dementia guide the development of multiple therapeutic approaches for AD and are the subject of this review.
Collapse
Affiliation(s)
- Donna M Barten
- Bristol Myers Squibb, Neuroscience Drug Discovery, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | |
Collapse
|
523
|
|
524
|
Kobayashi D, Zeller M, Cole T, Buttini M, McConlogue L, Sinha S, Freedman S, Morris RGM, Chen KS. BACE1 gene deletion: impact on behavioral function in a model of Alzheimer's disease. Neurobiol Aging 2008; 29:861-73. [PMID: 17331621 DOI: 10.1016/j.neurobiolaging.2007.01.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 11/15/2006] [Accepted: 01/13/2007] [Indexed: 10/23/2022]
Abstract
Accumulation of cerebral amyloid-beta (Abeta) has been implicated as a putative causal factor in the development of Alzheimer's disease (AD). Transgenic mice like the PDAPP line overexpress human mutant Amyloid Precursor Protein (hAPP) and recapitulate many features of AD, including amyloid neuropathology and cognitive deficits. Inhibition of the beta-site aspartyl cleaving enzyme (BACE1) enzyme responsible for the first proteolytic cleavage that ultimately generates Abeta has been proposed as a strategy for AD therapy. To assess the theoretical repercussions of beta-secretase activity reduction in an in vivo model of AD, BACE1(-/-) mice bred to the PDAPP line were examined in a series of behavioral tasks. Although BACE1 gene ablation abolished hAbeta accumulation, BACE1(-/-) mice had unexpected sensorimotor impairments, spatial memory deficits, and displayed seizures, phenotypes which were severe on the PDAPP background. These results suggest that while excess Abeta is functionally pathological, BACE1-mediated processing of APP and other substrates play a role in "normal" learning, memory and sensorimotor processes.
Collapse
Affiliation(s)
- Dione Kobayashi
- Rinat Neurosciences, 230 East Grand Avenue, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
525
|
Charrier N, Clarke B, Cutler L, Demont E, Dingwall C, Dunsdon R, East P, Hawkins J, Howes C, Hussain I, Jeffrey P, Maile G, Matico R, Mosley J, Naylor A, O’Brien A, Redshaw S, Rowland P, Soleil V, Smith KJ, Sweitzer S, Theobald P, Vesey D, Walter DS, Wayne G. Second Generation of Hydroxyethylamine BACE-1 Inhibitors: Optimizing Potency and Oral Bioavailability. J Med Chem 2008; 51:3313-7. [DOI: 10.1021/jm800138h] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Nicolas Charrier
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Brian Clarke
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Leanne Cutler
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Emmanuel Demont
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Colin Dingwall
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Rachel Dunsdon
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Philip East
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Julie Hawkins
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Colin Howes
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Ishrut Hussain
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Phil Jeffrey
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Graham Maile
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Rosalie Matico
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Julie Mosley
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Alan Naylor
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Alistair O’Brien
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Sally Redshaw
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Paul Rowland
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Virginie Soleil
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Kathrine J. Smith
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Sharon Sweitzer
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Pam Theobald
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - David Vesey
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Daryl S. Walter
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| | - Gareth Wayne
- Neurology and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Park, Harlow, Essex, CM19 5AW, U.K
| |
Collapse
|
526
|
Iwata N, Higuchi M, Saido TC. [Therapeutic strategy by manipulating some protease activities for Alzheimer's disease]. Nihon Yakurigaku Zasshi 2008; 131:320-325. [PMID: 18480559 DOI: 10.1254/fpj.131.320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
527
|
Hu X, He W, Diaconu C, Tang X, Kidd GJ, Macklin WB, Trapp BD, Yan R. Genetic deletion of BACE1 in mice affects remyelination of sciatic nerves. FASEB J 2008; 22:2970-80. [PMID: 18413858 DOI: 10.1096/fj.08-106666] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACE1 is a promising therapeutic and preventive target for Alzheimer's disease because it is essential for amyloid deposition. However, the recent demonstration of BACE1 in modulating developmental myelination in both peripheral and central nervous systems raises a concern of its effect on myelin maintenance or remyelination, and inhibition of these processes will potentially be detrimental to the BACE1 inhibitor users who are susceptible to myelination diseases such as adult peripheral nerve injury or multiple sclerosis. In this report, we investigated the role of BACE1 during peripheral nerve remyelination in wild-type (WT) and BACE1-null mice. We show here that genetic deletion of BACE1 affects sciatic nerve remyelination. The impaired remyelination appears to stem from the loss of neuregulin-1 cleavage by BACE1. To demonstrate a direct cleavage of neuregulin-1 by BACE1, we have identified a BACE1 cleavage site that turns out be highly conserved among neuregulin-1 paralogues. Moreover, we show that neuregulin-1 family member neuregulin-3 is also cleavable by BACE1. We hypothesize that the BACE1-cleaved extracellular domain of axonal neuregulin-1, perhaps neuregulin-3 as well, binds to Schwann cell ErbB receptors, which in turn regulate remyelination. Pharmacological inhibition of BACE1 should be carefully monitored to avoid alteration of signaling pathway that regulates remyelination.
Collapse
Affiliation(s)
- Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
528
|
Buggia-Prevot V, Sevalle J, Rossner S, Checler F. NFκB-dependent Control of BACE1 Promoter Transactivation by Aβ42. J Biol Chem 2008; 283:10037-47. [DOI: 10.1074/jbc.m706579200] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
529
|
Wen Y, Yu WH, Maloney B, Bailey J, Ma J, Marié I, Maurin T, Wang L, Figueroa H, Herman M, Krishnamurthy P, Liu L, Planel E, Lau LF, Lahiri DK, Duff K. Transcriptional regulation of beta-secretase by p25/cdk5 leads to enhanced amyloidogenic processing. Neuron 2008; 57:680-90. [PMID: 18341989 PMCID: PMC2329816 DOI: 10.1016/j.neuron.2008.02.024] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 01/10/2008] [Accepted: 02/19/2008] [Indexed: 12/22/2022]
Abstract
Cyclin-dependent kinase 5 (cdk5) has been implicated in Alzheimer's disease (AD) pathogenesis. Here, we demonstrate that overexpression of p25, an activator of cdk5, led to increased levels of BACE1 mRNA and protein in vitro and in vivo. A p25/cdk5 responsive region containing multiple sites for signal transducer and activator of transcription (STAT1/3) was identified in the BACE1 promoter. STAT3 interacts with the BACE1 promoter, and p25-overexpressing mice had elevated levels of pSTAT3 and BACE1, whereas cdk5-deficient mice had reduced levels. Furthermore, mice with a targeted mutation in the STAT3 cdk5 responsive site had lower levels of BACE1. Increased BACE levels in p25 overexpressing mice correlated with enhanced amyloidogenic processing that could be reversed by a cdk5 inhibitor. These data demonstrate a pathway by which p25/cdk5 increases the amyloidogenic processing of APP through STAT3-mediated transcriptional control of BACE1 that could have implications for AD pathogenesis.
Collapse
Affiliation(s)
- Yi Wen
- Taub Institute at Columbia University Medical Center, New York, NY
| | - W. Haung Yu
- Taub Institute at Columbia University Medical Center, New York, NY
| | - Bryan Maloney
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - Jason Bailey
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - Junrong Ma
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - Isabelle Marié
- Department of Pathology, New York University School of Medicine
| | - Thomas Maurin
- Taub Institute at Columbia University Medical Center, New York, NY
| | - Lili Wang
- Taub Institute at Columbia University Medical Center, New York, NY
| | - Helen Figueroa
- Taub Institute at Columbia University Medical Center, New York, NY
| | - Mathieu Herman
- Taub Institute at Columbia University Medical Center, New York, NY
| | | | - Li Liu
- Taub Institute at Columbia University Medical Center, New York, NY
| | - Emmanuel Planel
- Taub Institute at Columbia University Medical Center, New York, NY
| | - Lit-Fui Lau
- CNS Discovery, Pfizer Global Research and Development, Groton, CT
| | - Debomoy K. Lahiri
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - Karen Duff
- Taub Institute at Columbia University Medical Center, New York, NY
| |
Collapse
|
530
|
Small DH, Klaver DW, Beckman M. Regulation of proBACE1 by Glycosaminoglycans. NEURODEGENER DIS 2008; 5:206-8. [DOI: 10.1159/000113703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
531
|
Sankaranarayanan S, Price EA, Wu G, Crouthamel MC, Shi XP, Tugusheva K, Tyler KX, Kahana J, Ellis J, Jin L, Steele T, Stachel S, Coburn C, Simon AJ. In vivo beta-secretase 1 inhibition leads to brain Abeta lowering and increased alpha-secretase processing of amyloid precursor protein without effect on neuregulin-1. J Pharmacol Exp Ther 2008; 324:957-69. [PMID: 18156464 DOI: 10.1124/jpet.107.130039] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
beta-Secretase (BACE) cleavage of amyloid precursor protein (APP) is one of the first steps in the production of amyloid beta peptide Abeta42, the putative neurotoxic species in Alzheimer's disease. Recent studies have shown that BACE1 knockdown leads to hypomyelination, putatively caused by a decline in neuregulin (NRG)-1 processing. In this study, we have tested a potent cell-permeable BACE1 inhibitor (IC(50) approximately 30 nM) by administering it directly into the lateral ventricles of mice, expressing human wild-type (WT)-APP, to determine the consequences of BACE1 inhibition on brain APP and NRG-1 processing. BACE1 inhibition, in vivo, led to a significant dose- and time-dependent lowering of brain Abeta40 and Abeta42. BACE1 inhibition also led to a robust brain secreted (s)APPbeta lowering that was accompanied by an increase in brain sAPPalpha levels. Although an increase in full-length NRG-1 levels was evident in 15-day-old BACE1 homozygous knockout (KO) (-/-) mice, in agreement with previous studies, this effect was also observed in 15-day-old heterozygous (+/-) mice, but it was not evident in 30-day-old and 2-year-old BACE1 KO (-/-) mice. Thus, BACE1 knockdown led to a transient decrease in NRG-1 processing in mice. Pharmacological inhibition of BACE1 in adult mice, which led to significant Abeta lowering, was without any significant effect on brain NRG-1 processing. Taken together, these results suggest that BACE1 is the major beta-site cleavage enzyme for APP and that its inhibition can lower brain Abeta and redirect APP processing via the potentially nonamyloidogenic alpha-secretase pathway, without significantly altering NRG-1 processing.
Collapse
Affiliation(s)
- Sethu Sankaranarayanan
- Department of Alzheimer's Research, WP 26A-2000, Merck Research Laboratories, 770, Sumneytown Pike, West Point, PA 19486, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
532
|
Miller-Fleming L, Giorgini F, Outeiro TF. Yeast as a model for studying human neurodegenerative disorders. Biotechnol J 2008; 3:325-38. [PMID: 18228539 DOI: 10.1002/biot.200700217] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Protein misfolding and aggregation are central events in many disorders including several neurodegenerative diseases. This suggests that alterations in normal protein homeostasis may contribute to pathogenesis, but the exact molecular mechanisms involved are still poorly understood. The budding yeast Saccharomyces cerevisiae is one of the model systems of choice for studies in molecular medicine. Modeling human neurodegenerative diseases in this simple organism has already shown the incredible power of yeast to unravel the complex mechanisms and pathways underlying these pathologies. Indeed, this work has led to the identification of several potential therapeutic targets and drugs for many diseases, including the neurodegenerative diseases. Several features associated with these diseases, such as formation of protein aggregates, cellular toxicity mediated by misfolded proteins, oxidative stress and hallmarks of apoptosis have been faithfully recapitulated in yeast, enabling researchers to take advantage of this powerful model to rapidly perform genetic and compound screens with the aim of identifying novel candidate therapeutic targets and drugs. Here we review the work undertaken to model human brain disorders in yeast, and how these models provide insight into novel therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Leonor Miller-Fleming
- Instituto de Medicina Molecular, Cellular and Molecular Neuroscience Unit, Instituto de Fisiologia, Facultade [corrected] de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | | | | |
Collapse
|
533
|
Salloway S, Mintzer J, Weiner MF, Cummings JL. Disease-modifying therapies in Alzheimer's disease. Alzheimers Dement 2008; 4:65-79. [PMID: 18631951 DOI: 10.1016/j.jalz.2007.10.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Accepted: 10/24/2007] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a chronic, progressive, neurodegenerative disorder that places a substantial burden on patients, their families, and society. The disease affects approximately 5 million individuals in the United States, with an annual cost of care greater than $100 billion. During the past dozen years, several agents have been approved that enhance cognition and global function of AD patients, and recent advances in understanding AD pathogenesis has led to the development of numerous compounds that might modify the disease process. A wide array of antiamyloid and neuroprotective therapeutic approaches are under investigation on the basis of the hypothesis that amyloid beta (A beta) protein plays a pivotal role in disease onset and progression and that secondary consequences of A beta generation and deposition, including tau hyperphosphorylation and neurofibrillary tangle formation, oxidation, inflammation, and excitotoxicity, contribute to the disease process. Interventions in these processes with agents that reduce amyloid production, limit aggregation, or increase removal might block the cascade of events comprising AD pathogenesis. Reducing tau hyperphosphorylation, limiting oxidation and excitotoxicity, and controlling inflammation might be beneficial disease-modifying strategies. Potentially neuroprotective and restorative treatments such as neurotrophins, neurotrophic factor enhancers, and stem cell-related approaches are also under investigation.
Collapse
Affiliation(s)
- Stephen Salloway
- Department of Clinical Neuroscience, Division of Biology and Medicine, Brown Medical School, Providence, RI, USA.
| | | | | | | |
Collapse
|
534
|
Clarke B, Demont E, Dingwall C, Dunsdon R, Faller A, Hawkins J, Hussain I, MacPherson D, Maile G, Matico R, Milner P, Mosley J, Naylor A, O’Brien A, Redshaw S, Riddell D, Rowland P, Soleil V, Smith KJ, Stanway S, Stemp G, Sweitzer S, Theobald P, Vesey D, Walter DS, Ward J, Wayne G. BACE-1 inhibitors Part 1: Identification of novel hydroxy ethylamines (HEAs). Bioorg Med Chem Lett 2008; 18:1011-6. [DOI: 10.1016/j.bmcl.2007.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 12/07/2007] [Accepted: 12/11/2007] [Indexed: 10/22/2022]
|
535
|
La maladie d’Alzheimer : des lésions cérébrales aux perspectives thérapeutiques. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2008. [DOI: 10.1016/s0001-4079(19)32836-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
536
|
Hook VYH, Kindy M, Hook G. Inhibitors of cathepsin B improve memory and reduce beta-amyloid in transgenic Alzheimer disease mice expressing the wild-type, but not the Swedish mutant, beta-secretase site of the amyloid precursor protein. J Biol Chem 2008; 283:7745-53. [PMID: 18184658 DOI: 10.1074/jbc.m708362200] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Elucidation of Abeta-lowering agents that inhibit processing of the wild-type (WT) beta-secretase amyloid precursor protein (APP) site, present in most Alzheimer disease (AD) patients, is a logical approach for improving memory deficit in AD. The cysteine protease inhibitors CA074Me and E64d were selected by inhibition of beta-secretase activity in regulated secretory vesicles that produce beta-amyloid (Abeta). The regulated secretory vesicle activity, represented by cathepsin B, selectively cleaves the WT beta-secretase site but not the rare Swedish mutant beta-secretase site. In vivo treatment of London APP mice, expressing the WT beta-secretase site, with these inhibitors resulted in substantial improvement in memory deficit assessed by the Morris water maze test. After inhibitor treatment, the improved memory function was accompanied by reduced amyloid plaque load, decreased Abeta40 and Abeta42, and reduced C-terminal beta-secretase fragment derived from APP by beta-secretase. However, the inhibitors had no effects on any of these parameters in mice expressing the Swedish mutant beta-secretase site of APP. The notable efficacy of these inhibitors to improve memory and reduce Abeta in an AD animal model expressing the WT beta-secretase APP site present in the majority of AD patients provides support for CA074Me and E64d inhibitors as potential AD therapeutic agents.
Collapse
Affiliation(s)
- Vivian Y H Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA.
| | | | | |
Collapse
|
537
|
|
538
|
PI3-K- and PKC-dependent up-regulation of APP processing enzymes by retinoic acid. Biochem Biophys Res Commun 2008; 365:298-303. [DOI: 10.1016/j.bbrc.2007.10.167] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 10/28/2007] [Indexed: 11/19/2022]
|
539
|
Duyckaerts C, Potier MC, Delatour B. Alzheimer disease models and human neuropathology: similarities and differences. Acta Neuropathol 2008; 115:5-38. [PMID: 18038275 PMCID: PMC2100431 DOI: 10.1007/s00401-007-0312-8] [Citation(s) in RCA: 310] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2007] [Revised: 10/13/2007] [Accepted: 10/14/2007] [Indexed: 12/02/2022]
Abstract
Animal models aim to replicate the symptoms, the lesions or the cause(s) of Alzheimer disease. Numerous mouse transgenic lines have now succeeded in partially reproducing its lesions: the extracellular deposits of Abeta peptide and the intracellular accumulation of tau protein. Mutated human APP transgenes result in the deposition of Abeta peptide, similar but not identical to the Abeta peptide of human senile plaque. Amyloid angiopathy is common. Besides the deposition of Abeta, axon dystrophy and alteration of dendrites have been observed. All of the mutations cause an increase in Abeta 42 levels, except for the Arctic mutation, which alters the Abeta sequence itself. Overexpressing wild-type APP alone (as in the murine models of human trisomy 21) causes no Abeta deposition in most mouse lines. Doubly (APP x mutated PS1) transgenic mice develop the lesions earlier. Transgenic mice in which BACE1 has been knocked out or overexpressed have been produced, as well as lines with altered expression of neprilysin, the main degrading enzyme of Abeta. The APP transgenic mice have raised new questions concerning the mechanisms of neuronal loss, the accumulation of Abeta in the cell body of the neurons, inflammation and gliosis, and the dendritic alterations. They have allowed some insight to be gained into the kinetics of the changes. The connection between the symptoms, the lesions and the increase in Abeta oligomers has been found to be difficult to unravel. Neurofibrillary tangles are only found in mouse lines that overexpress mutated tau or human tau on a murine tau -/- background. A triply transgenic model (mutated APP, PS1 and tau) recapitulates the alterations seen in AD but its physiological relevance may be discussed. A number of modulators of Abeta or of tau accumulation have been tested. A transgenic model may be analyzed at three levels at least (symptoms, lesions, cause of the disease), and a reading key is proposed to summarize this analysis.
Collapse
Affiliation(s)
- Charles Duyckaerts
- Laboratoire de Neuropathologie Raymond Escourolle, Hôpital de La Salpêtrière, 47 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France.
| | | | | |
Collapse
|
540
|
|
541
|
Spencer B, Rockenstein E, Crews L, Marr R, Masliah E. Novel strategies for Alzheimer's disease treatment. Expert Opin Biol Ther 2007; 7:1853-67. [PMID: 18034651 DOI: 10.1517/14712598.7.12.1853] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Considerable progress has been made in recent years towards better understanding the pathogenesis of Alzheimer's disease (AD), a dementing neurodegenerative disorder that affects > 10 million individuals in the US and Europe combined. Recent studies suggest that alterations in the processing of amyloid precursor protein (APP), resulting in the accumulation of amyloid-beta protein (Abeta) and the formation of oligomers leads to synaptic damage and neurodegeneration. Therefore, strategies for treatment development have been focused on reducing Abeta accumulation using, among other approaches, antiaggregation molecules, regulators of the APP proteolysis and processing, reducing APP production (e.g., small-interfering RNA), and increasing Abeta clearance with antibodies, apolipoprotein E and Abeta-degrading enzymes (e.g., neprilysin). The main focus of this review is on novel treatments for AD with a special emphasis on delivering neuroprotective and antiamyloidogenic molecules by gene therapy and by promoting neurogenesis.
Collapse
Affiliation(s)
- Brian Spencer
- University of California, Department of Neurosciences, San Diego, La Jolla, CA 92093-0624, USA
| | | | | | | | | |
Collapse
|
542
|
Sugimoto I, Futakawa S, Oka R, Ogawa K, Marth JD, Miyoshi E, Taniguchi N, Hashimoto Y, Kitazume S. Beta-galactoside alpha2,6-sialyltransferase I cleavage by BACE1 enhances the sialylation of soluble glycoproteins. A novel regulatory mechanism for alpha2,6-sialylation. J Biol Chem 2007; 282:34896-34903. [PMID: 17897958 DOI: 10.1074/jbc.m704766200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACE1 (beta-site amyloid precursor protein-cleaving enzyme-1) is a membrane-bound aspartic protease that cleaves amyloid precursor protein to produce a neurotoxic peptide, amyloid beta-peptide, and has been implicated in triggering the pathogenesis of Alzheimer disease. We showed previously that BACE1 cleaves beta-galactoside alpha2,6-sialyltransferase I (ST6Gal I) to initiate its secretion, but it remained unclear how BACE1 affects the cellular level of alpha2,6-sialylation. Here, we found that BACE1 overexpression in Hep3B cells increased the sialylation of soluble secreted glycoproteins, but did not affect cell-surface sialylation. The sialylation of soluble glycoproteins was not increased by ST6Gal I overexpression alone, but was increased by co-overexpression of ST6Gal I and BACE1 or by expression of the soluble form of ST6Gal I, suggesting that soluble ST6Gal I produced by BACE1 plays, at least in part, a role in the sialylation of soluble glycoproteins. We also found that plasma glycoproteins from BACE1-deficient mice exhibited reduced levels of alpha2,6-sialylation compared with those from wild-type mice. We propose a novel regulatory mechanism in which cleavage and secretion of ST6Gal I enhance the sialylation of soluble glycoprotein substrates.
Collapse
Affiliation(s)
- Ichiro Sugimoto
- Glyco-Chain Functions Laboratory, Institute of Physical and Chemical Research, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
543
|
Cole SL, Vassar R. The Alzheimer's disease beta-secretase enzyme, BACE1. Mol Neurodegener 2007; 2:22. [PMID: 18005427 PMCID: PMC2211305 DOI: 10.1186/1750-1326-2-22] [Citation(s) in RCA: 374] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Accepted: 11/15/2007] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of Alzheimer's disease is highly complex. While several pathologies characterize this disease, amyloid plaques, composed of the β-amyloid peptide are hallmark neuropathological lesions in Alzheimer's disease brain. Indeed, a wealth of evidence suggests that β-amyloid is central to the pathophysiology of AD and is likely to play an early role in this intractable neurodegenerative disorder. The BACE1 enzyme is essential for the generation of β-amyloid. BACE1 knockout mice do not produce β-amyloid and are free from Alzheimer's associated pathologies including neuronal loss and certain memory deficits. The fact that BACE1 initiates the formation of β-amyloid, and the observation that BACE1 levels are elevated in this disease provide direct and compelling reasons to develop therapies directed at BACE1 inhibition thus reducing β-amyloid and its associated toxicities. However, new data indicates that complete abolishment of BACE1 may be associated with specific behavioral and physiological alterations. Recently a number of non-APP BACE1 substrates have been identified. It is plausible that failure to process certain BACE1 substrates may underlie some of the reported abnormalities in the BACE1-deficient mice. Here we review BACE1 biology, covering aspects ranging from the initial identification and characterization of this enzyme to recent data detailing the apparent dysregulation of BACE1 in Alzheimer's disease. We pay special attention to the putative function of BACE1 during healthy conditions and discuss in detail the relationship that exists between key risk factors for AD, such as vascular disease (and downstream cellular consequences), and the pathogenic alterations in BACE1 that are observed in the diseased state.
Collapse
Affiliation(s)
- Sarah L Cole
- Department of Cell and Molecular Biology, The Feinberg School of Medicine, Northwestern University, Chicago Avenue, Chicago, IL, USA.
| | | |
Collapse
|
544
|
|
545
|
Liu Q, Zerbinatti CV, Zhang J, Hoe HS, Wang B, Cole SL, Herz J, Muglia L, Bu G. Amyloid precursor protein regulates brain apolipoprotein E and cholesterol metabolism through lipoprotein receptor LRP1. Neuron 2007; 56:66-78. [PMID: 17920016 PMCID: PMC2045076 DOI: 10.1016/j.neuron.2007.08.008] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 07/06/2007] [Accepted: 08/13/2007] [Indexed: 12/13/2022]
Abstract
Mutations in the amyloid precursor protein (APP) cause early-onset Alzheimer's disease (AD), but the only genetic risk factor for late-onset AD is the varepsilon4 allele of apolipoprotein E (apoE), a major cholesterol carrier. Using Cre-lox conditional knockout mice, we demonstrate that lipoprotein receptor LRP1 expression regulates apoE and cholesterol levels within the CNS. We also found that deletion of APP and its homolog APLP2, or components of the gamma-secretase complex, significantly enhanced the expression and function of LRP1, which was reversed by forced expression of the APP intracellular domain (AICD). We further show that AICD, together with Fe65 and Tip60, interacts with the LRP1 promoter and suppresses its transcription. Together, our findings support that the gamma-secretase cleavage of APP plays a central role in regulating apoE and cholesterol metabolism in the CNS via LRP1 and establish a biological linkage between APP and apoE, the two major genetic determinants of AD.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
546
|
McConlogue L, Buttini M, Anderson JP, Brigham EF, Chen KS, Freedman SB, Games D, Johnson-Wood K, Lee M, Zeller M, Liu W, Motter R, Sinha S. Partial reduction of BACE1 has dramatic effects on Alzheimer plaque and synaptic pathology in APP Transgenic Mice. J Biol Chem 2007; 282:26326-34. [PMID: 17616527 DOI: 10.1074/jbc.m611687200] [Citation(s) in RCA: 242] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The aspartyl protease beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) initiates processing of amyloid precursor protein (APP) into amyloid beta (Abeta) peptide, the major component of Alzheimer disease (AD) plaques. To determine the role that BACE1 plays in the development of Abeta-driven AD-like pathology, we have crossed PDAPP mice, a transgenic mouse model of AD overexpressing human mutated APP, onto mice with either a homozygous or heterozygous BACE1 gene knockout. Analysis of PDAPP/BACE(-/-) mice demonstrated that BACE1 is absolutely required for both Abeta generation and the development of age-associated plaque pathology. Furthermore, synaptic deficits, a neurodegenerative pathology characteristic of AD, were also reversed in the bigenic mice. To determine the extent of BACE1 reduction required to significantly inhibit pathology, PDAPP mice having a heterozygous BACE1 gene knock-out were evaluated for Abeta generation and for the development of pathology. Although the 50% reduction in BACE1 enzyme levels caused only a 12% decrease in Abeta levels in young mice, it nonetheless resulted in a dramatic reduction in Abeta plaques, neuritic burden, and synaptic deficits in older mice. Quantitative analyses indicate that brain Abeta levels in young APP transgenic mice are not the sole determinant for the changes in plaque pathology mediated by reduced BACE1. These observations demonstrate that partial reductions of BACE1 enzyme activity and concomitant Abeta levels lead to dramatic inhibition of Abeta-driven AD-like pathology, making BACE1 an excellent target for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Lisa McConlogue
- Department of Biology, Elan Pharmaceuticals, South San Francisco, California 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
547
|
Hiraoka Y, Ohno M, Yoshida K, Okawa K, Tomimoto H, Kita T, Nishi E. Enhancement of alpha-secretase cleavage of amyloid precursor protein by a metalloendopeptidase nardilysin. J Neurochem 2007; 102:1595-1605. [PMID: 17555553 DOI: 10.1111/j.1471-4159.2007.04685.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Amyloid-beta (Abeta) peptide, the principal component of senile plaques in the brains of patients with Alzheimer's disease, is derived from proteolytic cleavage of amyloid precursor protein (APP) by beta- and gamma-secretases. Alternative cleavage of APP by alpha-secretase occurs within the Abeta domain and precludes generation of Abeta peptide. Three members of the ADAM (a disintegrin and metalloprotease) family of proteases, ADAM9, 10 and 17, are the main candidates for alpha-secretases. However, the mechanism that regulates alpha-secretase activity remains unclear. We have recently demonstrated that nardilysin (EC 3.4.24.61, N-arginine dibasic convertase; NRDc) enhances ectodomain shedding of heparin-binding epidermal growth factor-like growth factor through activation of ADAM17. In this study, we show that NRDc enhances the alpha-secretase activity of ADAMs, which results in a decrease in the amount of Abeta generated. When expressed with ADAMs in cells, NRDc dramatically increased the secretion of alpha-secretase-cleaved soluble APP and reduced the amount of Abeta peptide generated. A peptide cleavage assay in vitro also showed that recombinant NRDc enhances ADAM17-induced cleavage of the peptide substrate corresponding to the alpha-secretase cleavage site of APP. A reduction of endogenous NRDc by RNA interference was accompanied by a decrease in the cleavage by alpha-secretase of APP and increase in the amount of Abeta generated. Notably, NRDc is clearly expressed in cortical neurons in human brain. Our results indicate that NRDc is involved in the metabolism of APP through regulation of the alpha-secretase activity of ADAMs, which may be a novel target for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yoshinori Hiraoka
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mikiko Ohno
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Yoshida
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuya Okawa
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidekazu Tomimoto
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toru Kita
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eiichiro Nishi
- Molecular Pathology Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, JapanBiomolecular Characterization Unit, Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto, JapanDepartment of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
548
|
Chang WP, Downs D, Huang XP, Da H, Fung KM, Tang J. Amyloid‐beta reduction by memapsin 2 (beta‐secretase) immunization. FASEB J 2007; 21:3184-96. [PMID: 17494994 DOI: 10.1096/fj.06-7993com] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Memapsin 2 (beta-secretase, BACE1) is the protease that initiates cleavage of beta-amyloid precursor protein leading to the production of amyloid-beta (Abeta) and the onset of Alzheimer's disease (AD). Reducing Abeta by targeting memapsin 2 is a major strategy in developing new AD therapy. Here, in a proof-of-concept study, we show that immunization of transgenic AD mice (Tg2576) with memapsin 2 resulted in Abeta reduction and cognitive improvement. To study the basis of this therapy, we demonstrated that anti-memapsin 2 (anti-M2) antibodies were rapidly internalized and reduced Abeta production in cultured cells. These antibodies also effectively crossed the blood-brain barrier to reach the brain. Two- and 10-month Tg2576 mice were immunized and monitored over 10 and 6 months, respectively. We observed a significant decrease of plasma and brain Abeta40 and Abeta42 (approximately 35%) in the immunized mice as compared to controls. Immunized mice also showed better cognitive performance than controls in both cohorts. Brain histological analyses found no evidence of T cell/microglia/astrocyte activation in the immunized mice, suggesting the absence of inflammatory responses. These results suggest that memapsin 2 immunization in Tg2576 was effective in reducing Abeta production and improving cognitive function and that the current approach warrants further investigation as a therapy for AD.
Collapse
Affiliation(s)
- Wan-Pin Chang
- Protein Studies Program, Oklahoma Medical Research Foundation, 825 N.E. 13th St., Oklahoma City, OK 73104, USA.
| | | | | | | | | | | |
Collapse
|
549
|
Zhao J, Fu Y, Yasvoina M, Shao P, Hitt B, O'Connor T, Logan S, Maus E, Citron M, Berry R, Binder L, Vassar R. Beta-site amyloid precursor protein cleaving enzyme 1 levels become elevated in neurons around amyloid plaques: implications for Alzheimer's disease pathogenesis. J Neurosci 2007; 27:3639-49. [PMID: 17409228 PMCID: PMC6672403 DOI: 10.1523/jneurosci.4396-06.2007] [Citation(s) in RCA: 301] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) (beta-secretase) initiates generation of beta-amyloid (Abeta), which plays an early role in Alzheimer's disease (AD). BACE1 levels are increased in postmortem AD brain, suggesting BACE1 elevation promotes Abeta production and AD. Alternatively, the BACE1 increase may be an epiphenomenon of late-stage AD. To distinguish between these possibilities, we analyzed BACE1 elevation using a highly specific BACE1 antibody, BACE-Cat1, made in BACE1-/- mice, which mount a robust anti-BACE1 immune response. Previous BACE1 immunohistochemical studies lack consistent results because typical BACE1 antibodies produce nonspecific background, but BACE-Cat1 immunolabels BACE1 only. BACE1 elevation was recapitulated in two amyloid precursor protein (APP) transgenic mouse lines. 5XFAD mice form amyloid plaques at young ages and exhibit neuron loss. In contrast, Tg2576 form plaques at a more advanced age and do not show cell death. These two mouse lines allow differentiation between early Abeta-induced events and late phenomena related to neuron death. BACE1 levels became elevated in parallel with amyloid burden in each APP transgenic, starting early in 5XFAD and late in Tg2576. The increase in BACE1 protein occurred without any change in BACE1 mRNA level, indicating a posttranscriptional mechanism. In APP transgenic and AD brains, high BACE1 levels were observed in an annulus around Abeta42-positive plaque cores and colocalized with neuronal proteins. These results demonstrate that amyloid plaques induce BACE1 in surrounding neurons at early stages of pathology before neuron death occurs. We conclude that BACE1 elevation is most likely triggered by the amyloid pathway and may drive a positive-feedback loop in AD.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Yifan Fu
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Marina Yasvoina
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Peizhen Shao
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Brian Hitt
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Tracy O'Connor
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Sreemathi Logan
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Erika Maus
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | | | - Robert Berry
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Lester Binder
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Robert Vassar
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| |
Collapse
|
550
|
Ma H, Lesné S, Kotilinek L, Steidl-Nichols JV, Sherman M, Younkin L, Younkin S, Forster C, Sergeant N, Delacourte A, Vassar R, Citron M, Kofuji P, Boland LM, Ashe KH. Involvement of beta-site APP cleaving enzyme 1 (BACE1) in amyloid precursor protein-mediated enhancement of memory and activity-dependent synaptic plasticity. Proc Natl Acad Sci U S A 2007; 104:8167-72. [PMID: 17470798 PMCID: PMC1859992 DOI: 10.1073/pnas.0609521104] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The amyloid precursor protein (APP) undergoes sequential cleavages to generate various polypeptides, including the amyloid-beta protein (Abeta), which forms amyloid plaques in Alzheimer's disease (AD), secreted APPalpha (sAPPalpha) which enhances memory, and the APP intracellular domain (AICD), which has been implicated in the regulation of gene transcription and calcium signaling. The beta-site APP cleaving enzyme 1 (BACE1) cleaves APP in an activity-dependent manner to form Abeta, AICD, and secreted APPbeta. Because this neural activity was shown to diminish synaptic transmission in vitro [Kamenetz F, Tomita T, Hsieh H, Seabrook G, Borchelt D, Iwatsubo T, Sisodia S, Malinow R (2003) Neuron 37:925-937], the prevailing notion has been that this pathway diminishes synaptic function. Here we investigated the role of this pathway in vivo. We studied transgenic mice overproducing APP that do not develop AD pathology or memory deficits but instead exhibit enhanced spatial memory. We showed enhanced synaptic plasticity in the hippocampus that depends on prior synaptic activity. We found that the enhanced memory and synaptic plasticity are abolished by the ablation of one or both copies of the BACE1 gene, leading to a significant decrease in AICD but not of any other APP cleavage products. In contrast to the previously described negative effect of BACE1-mediated cleavage of APP on synaptic function in vitro, our in vivo work indicates that BACE1-mediated cleavage of APP can facilitate learning, memory, and synaptic plasticity.
Collapse
Affiliation(s)
- Huifang Ma
- Departments of Neurology
- Neuroscience, and
| | | | | | | | | | - Linda Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - Steven Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - Colleen Forster
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Nicolas Sergeant
- Institut National de la Santé et de la Recherche Médicale U837, Centre Jean Pierre Aubert, F-59045 Lille, France
| | - André Delacourte
- Institut National de la Santé et de la Recherche Médicale U837, Centre Jean Pierre Aubert, F-59045 Lille, France
| | - Robert Vassar
- Department of Neuroscience, Northwestern University, Evanston, IL 60208
| | - Martin Citron
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320; and
| | | | | | - Karen H. Ashe
- Departments of Neurology
- Neuroscience, and
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Medical Center, Minneapolis, MN 55417
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|