351
|
Zhou L, Barão S, Laga M, Bockstael K, Borgers M, Gijsen H, Annaert W, Moechars D, Mercken M, Gevaert K, Gevaer K, De Strooper B. The neural cell adhesion molecules L1 and CHL1 are cleaved by BACE1 protease in vivo. J Biol Chem 2012; 287:25927-40. [PMID: 22692213 DOI: 10.1074/jbc.m112.377465] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The β-site amyloid precursor protein-cleaving enzyme BACE1 is a prime drug target for Alzheimer disease. However, the function and the physiological substrates of BACE1 remain largely unknown. In this work, we took a quantitative proteomic approach to analyze the secretome of primary neurons after acute BACE1 inhibition, and we identified several novel substrate candidates for BACE1. Many of these molecules are involved in neuronal network formation in the developing nervous system. We selected the adhesion molecules L1 and CHL1, which are crucial for axonal guidance and maintenance of neural circuits, for further validation as BACE1 substrates. Using both genetic BACE1 knock-out and acute pharmacological BACE1 inhibition in mice and cell cultures, we show that L1 and CHL1 are cleaved by BACE1 under physiological conditions. The BACE1 cleavage sites at the membrane-proximal regions of L1 (between Tyr(1086) and Glu(1087)) and CHL1 (between Gln(1061) and Asp(1062)) were determined by mass spectrometry. This work provides molecular insights into the function and the pathways in which BACE1 is involved, and it will help to predict or interpret possible side effects of BACE1 inhibitor drugs in current clinical trials.
Collapse
Affiliation(s)
- Lujia Zhou
- VIB Center for the Biology of Disease, KULeuven, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
352
|
Kumar AB, Anderson JM, Melendez AL, Manetsch R. Synthesis and structure-activity relationship studies of 1,3-disubstituted 2-propanols as BACE-1 inhibitors. Bioorg Med Chem Lett 2012; 22:4740-4. [PMID: 22727644 DOI: 10.1016/j.bmcl.2012.05.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 05/16/2012] [Accepted: 05/18/2012] [Indexed: 12/11/2022]
Abstract
A library of 1,3-disubstituted 2-propanols was synthesized and evaluated as low molecular weight probes for β-secretase inhibition. By screening a library of 121 1,3-disubstituted 2-propanol derivatives, we identified few compounds inhibiting the enzyme at low micromolar concentrations. The initial hits were optimized to yield a potent BACE-1 inhibitor exhibiting an IC(50) constant in the nanomolar range. Exploration of the pharmacological properties revealed that these small molecular inhibitors possessed a high selectivity over cathepsin D and desirable physicochemical properties beneficial to cross the blood-brain barrier.
Collapse
Affiliation(s)
- Arun Babu Kumar
- Department of Chemistry, University of South Florida, CHE 205, 4202 E. Fowler Ave, Tampa, FL 33620, USA
| | | | | | | |
Collapse
|
353
|
Sandgren V, Agback T, Johansson PO, Lindberg J, Kvarnström I, Samuelsson B, Belda O, Dahlgren A. Highly potent macrocyclic BACE-1 inhibitors incorporating a hydroxyethylamine core: design, synthesis and X-ray crystal structures of enzyme inhibitor complexes. Bioorg Med Chem 2012; 20:4377-89. [PMID: 22698785 DOI: 10.1016/j.bmc.2012.05.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 04/17/2012] [Accepted: 05/16/2012] [Indexed: 11/30/2022]
Abstract
A series of P1-P3 linked macrocyclic BACE-1 inhibitors containing a hydroxyethylamine (HEA) isostere scaffold has been synthesized. All inhibitors comprise a toluene or N-phenylmethanesulfonamide P2 moiety. Excellent BACE-1 potencies, both in enzymatic and cell-based assays, were observed in this series of target compounds, with the best candidates displaying cell-based IC(50) values in the low nanomolar range. As an attempt to improve potency, a phenyl substituent aiming at the S3 subpocket was introduced in the macrocyclic ring. X-ray analyzes were performed on selected compounds, and enzyme-inhibitor interactions are discussed.
Collapse
Affiliation(s)
- Veronica Sandgren
- Department of Chemistry, Linköping University, S-581 83 Linköping, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
354
|
Barman A, Prabhakar R. Protonation states of the catalytic dyad of β-secretase (BACE1) in the presence of chemically diverse inhibitors: a molecular docking study. J Chem Inf Model 2012; 52:1275-87. [PMID: 22545704 DOI: 10.1021/ci200611t] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this molecular docking study, the protonation states of the catalytic Asp dyad of the beta-secretase (BACE1) enzyme in the presence of eight chemically diverse inhibitors have been predicted. BACE1 catalyzes the rate-determining step in the generation of Alzheimer amyloid beta peptides and is widely considered as a promising therapeutic target. All the inhibitors were redocked into their corresponding X-ray structures using a combination of eight different protonation states of the Asp dyad for each inhibitor. Five inhibitors were primarily found to favor two different monoprotonated states, and the remaining three favor a dideprotonated state. In addition, five of them exhibited secondary preference for a diprotonated state. These results show that the knowledge of a single protonation state of the Asp dyad is not sufficient to search for the novel inhibitors of BACE1 and the most plausible state for each inhibitor must be determined prior to conducting in-silico screening.
Collapse
Affiliation(s)
- Arghya Barman
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, USA
| | | |
Collapse
|
355
|
Plisson F, Conte M, Khalil Z, Huang XC, Piggott AM, Capon RJ. Kinase Inhibitor Scaffolds against Neurodegenerative Diseases from a Southern Australian Ascidian,Didemnumsp. ChemMedChem 2012; 7:983-90. [DOI: 10.1002/cmdc.201200169] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Indexed: 11/06/2022]
|
356
|
Probst G, Xu YZ. Small-molecule BACE1 inhibitors: a patent literature review (2006 - 2011). Expert Opin Ther Pat 2012; 22:511-40. [PMID: 22512789 DOI: 10.1517/13543776.2012.681302] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Alzheimer's disease is a devastating neurodegenerative disorder for which no disease-modifying therapy exists. The amyloid hypothesis, which implicates Aβ as the toxin initiating a biological cascade leading to neurodegeneration, is the most prominent theory concerning the underlying cause of the disease. BACE1 is one of two aspartyl proteinases that generate Aβ, thus inhibition of BACE1 has the potential to ameliorate the progression of Alzheimer's disease by abating the production of Aβ. AREAS COVERED This review chronicles small-molecule BACE1 inhibitors as described in the patent literature between 2006 and 2011 and their potential use as disease-modifying treatments for Alzheimer's disease. Over the past half a dozen years, numerous BACE1 inhibitors have been published in the patent applications, but often these contain a paltry amount of pertinent biological data (e.g. potency, selectivity, and efficacy). Fortunately, numerous relevant publications containing important data have appeared in the journal literature during this period. The goal in this effort was to create an amalgam of the two records to add value to this review. EXPERT OPINION The pharmaceutical industry has made tremendous progress in the development of small-molecule BACE1 inhibitors that lower Aβ in the central nervous system. Assuming the amyloid hypothesis is veracious, we anticipate a disease-modifying therapy to combat Alzheimer's disease is near.
Collapse
Affiliation(s)
- Gary Probst
- Elan Pharmaceuticals, Molecular Design, 180 Oyster Point Boulevard, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
357
|
Dineen TA, Weiss MM, Williamson T, Acton P, Babu-Khan S, Bartberger MD, Brown J, Chen K, Cheng Y, Citron M, Croghan MD, Dunn RT, Esmay J, Graceffa RF, Harried SS, Hickman D, Hitchcock SA, Horne DB, Huang H, Imbeah-Ampiah R, Judd T, Kaller MR, Kreiman CR, La DS, Li V, Lopez P, Louie S, Monenschein H, Nguyen TT, Pennington LD, San Miguel T, Sickmier EA, Vargas HM, Wahl RC, Wen PH, Whittington DA, Wood S, Xue Q, Yang BH, Patel VF, Zhong W. Design and Synthesis of Potent, Orally Efficacious Hydroxyethylamine Derived β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE1) Inhibitors. J Med Chem 2012; 55:9025-44. [DOI: 10.1021/jm300118s] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Thomas A. Dineen
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Matthew M. Weiss
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Toni Williamson
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Paul Acton
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Safura Babu-Khan
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Michael D. Bartberger
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - James Brown
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Kui Chen
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Yuan Cheng
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Martin Citron
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Michael D. Croghan
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Robert T. Dunn
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Joel Esmay
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Russell F. Graceffa
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Scott S. Harried
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Dean Hickman
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Stephen A. Hitchcock
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Daniel B. Horne
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Hongbing Huang
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Ronke Imbeah-Ampiah
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Ted Judd
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Matthew R. Kaller
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Charles R. Kreiman
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Daniel S. La
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Vivian Li
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Patricia Lopez
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Steven Louie
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Holger Monenschein
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Thomas T. Nguyen
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Lewis D. Pennington
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Tisha San Miguel
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - E. Allen Sickmier
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Hugo M. Vargas
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Robert C. Wahl
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Paul H. Wen
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Douglas A. Whittington
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Stephen Wood
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Qiufen Xue
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Bryant H. Yang
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Vinod F. Patel
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Wenge Zhong
- Chemical
Research and Discovery, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
- Department
of Neuroscience, §Department of HTS and Molecular Pharmacology, ∥Molecular Structure, ⊥Pharmacokinetics
and Drug Metabolism, #Comparative Biology and Safety Sciences, Amgen Inc., One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
358
|
Tayeb HO, Yang HD, Price BH, Tarazi FI. Pharmacotherapies for Alzheimer's disease: Beyond cholinesterase inhibitors. Pharmacol Ther 2012; 134:8-25. [DOI: 10.1016/j.pharmthera.2011.12.002] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 11/21/2011] [Indexed: 12/31/2022]
|
359
|
Rueeger H, Lueoend R, Rogel O, Rondeau JM, Möbitz H, Machauer R, Jacobson L, Staufenbiel M, Desrayaud S, Neumann U. Discovery of Cyclic Sulfone Hydroxyethylamines as Potent and Selective β-Site APP-Cleaving Enzyme 1 (BACE1) Inhibitors: Structure-Based Design and in Vivo Reduction of Amyloid β-Peptides. J Med Chem 2012; 55:3364-86. [DOI: 10.1021/jm300069y] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Heinrich Rueeger
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Rainer Lueoend
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Olivier Rogel
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Jean-Michel Rondeau
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Henrik Möbitz
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Rainer Machauer
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Laura Jacobson
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Matthias Staufenbiel
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Sandrine Desrayaud
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| | - Ulf Neumann
- Department
of Global Discovery Chemistry, ‡Structural Biology Platform, §Department of Neuroscience, and ∥Metabolism and
Pharmacokinetics, Institutes for BioMedical Research, Novartis Pharma AG, CH-4057 Basel, Switzerland
| |
Collapse
|
360
|
|
361
|
Alkadhi KA, Alzoubi KH, Srivareerat M, Tran TT. Elevation of BACE in an Aβ rat model of Alzheimer's disease: exacerbation by chronic stress and prevention by nicotine. Int J Neuropsychopharmacol 2012; 15:223-233. [PMID: 21356140 DOI: 10.1017/s1461145711000162] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
In Alzheimer's disease (AD), progressive accumulation of β-amyloid (Aβ) peptides impairs nicotinic acetylcholine receptor (nAChR) function by a mechanism that may involve α7 and α4β2-nAChR subtypes. Additionally, the beta-site amyloid precursor protein (APP)-cleaving enzyme (BACE), the rate-limiting enzyme in the pathogenic Aβ production pathway, is expressed at high levels in hippocampal and cortical regions of AD brains. We measured hippocampal area CA1 protein levels of BACE and α7- and α4β2-nAChR subunits using an Aβ rat model of AD (14-d osmotic pump i.c.v. infusion of 300 pmol/d Aβ peptides) in the presence and absence of chronic stress and/or chronic nicotine treatment. There was a significant increase in the levels of BACE in Aβ-infused rats, which were markedly intensified by chronic (4-6 wk) stress, but were normalized in Aβ rats chronically treated with nicotine (1 mg/kg b.i.d.). The levels of the three subunits α7, α4 and β2 were significantly decreased in Aβ rats, but these were also normalized in Aβ rats chronically treated with nicotine. Chronic stress did not further aggravate the reduction of nAChRs in Aβ-infused rats. The increased BACE levels and decreased nAChR levels, which are established hallmarks of AD, provide additional support for the validity of the Aβ i.c.v.-infused rat as a model of AD.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA.
| | | | | | | |
Collapse
|
362
|
Dislich B, Lichtenthaler SF. The Membrane-Bound Aspartyl Protease BACE1: Molecular and Functional Properties in Alzheimer's Disease and Beyond. Front Physiol 2012; 3:8. [PMID: 22363289 PMCID: PMC3281277 DOI: 10.3389/fphys.2012.00008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 01/11/2012] [Indexed: 12/31/2022] Open
Abstract
The β-site APP cleaving enzyme 1 (BACE1) is a transmembrane aspartyl protease involved in Alzheimer’s disease (AD) pathogenesis and in myelination. BACE1 initiates the generation of the pathogenic amyloid β-peptide, which makes BACE1 a major drug target for AD. BACE1 also cleaves and activates neuregulin 1, thereby contributing to postnatal myelination, in particular in the peripheral nervous system. Additional proteins are also cleaved by BACE1, but less is known about the physiological consequences of their cleavage. Recently, new phenotypes were described in BACE1-deficient mice. Although it remains unclear through which BACE1 substrates they are mediated, the phenotypes suggest a versatile role of this protease for diverse physiological processes. This review summarizes the enzymatic and cellular properties of BACE1 as well as its regulation by lipids, by transcriptional, and by translational mechanisms. The main focus will be on the recent progress in understanding BACE1 function and its implication for potential mechanism-based side effects upon therapeutic inhibition.
Collapse
Affiliation(s)
- Bastian Dislich
- German Center for Neurodegenerative Diseases (DZNE) Munich, Germany
| | | |
Collapse
|
363
|
Zhao Y, Wang Y, Hu J, Zhang X, Zhang YW. CutA divalent cation tolerance homolog (Escherichia coli) (CUTA) regulates β-cleavage of β-amyloid precursor protein (APP) through interacting with β-site APP cleaving protein 1 (BACE1). J Biol Chem 2012; 287:11141-50. [PMID: 22351782 DOI: 10.1074/jbc.m111.330209] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Accumulation of the neurotoxic β-amyloid (Aβ) peptide in the brain is central to the pathogenesis of Alzheimer disease. Aβ is derived from the β-amyloid precursor protein (APP) through sequential cleavages by β- and γ-secretases, and the production of Aβ is greatly affected by the subcellular localization of these factors. CUTA, the mammalian CutA divalent cation tolerance homolog (E. coli), has been proposed to mediate acetylcholinesterase activity and copper homeostasis, which are important in Alzheimer disease pathology. However, the exact function of CUTA remains largely unclear. Here we show that human CUTA has several variants that differ in their N-terminal length and are separated as heavy (H) and light (L) components. The H component has the longest N terminus and is membrane-associated, whereas the L component is N-terminally truncated at various sites and localized in the cytosol. Importantly, we demonstrate that the H component of CUTA interacts through its N terminus with the transmembrane domain of β-site APP cleaving enzyme 1 (BACE1), the putative β-secretase, mainly in the Golgi/trans-Golgi network. Overexpression and RNA interference knockdown of CUTA can reduce and increase BACE1-mediated APP processing/Aβ secretion, respectively. RNA interference of CUTA decelerates intracellular trafficking of BACE1 from the Golgi/trans-Golgi network to the cell surface and reduces the steady-state level of cell surface BACE1. Our results identify the H component of CUTA as a novel BACE1-interacting protein that mediates the intracellular trafficking of BACE1 and the processing of APP to Aβ.
Collapse
Affiliation(s)
- Yingjun Zhao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, College of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | | | | | | | | |
Collapse
|
364
|
Structure based design of iminohydantoin BACE1 inhibitors: identification of an orally available, centrally active BACE1 inhibitor. Bioorg Med Chem Lett 2012; 22:2444-9. [PMID: 22390835 DOI: 10.1016/j.bmcl.2012.02.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 02/03/2012] [Accepted: 02/06/2012] [Indexed: 11/21/2022]
Abstract
From an initial lead 1, a structure-based design approach led to identification of a novel, high-affinity iminohydantoin BACE1 inhibitor that lowers CNS-derived Aβ following oral administration to rats. Herein we report SAR development in the S3 and F' subsites of BACE1 for this series, the synthetic approaches employed in this effort, and in vivo data for the optimized compound.
Collapse
|
365
|
Zhang M, Deng Y, Luo Y, Zhang S, Zou H, Cai F, Wada K, Song W. Control of BACE1 degradation and APP processing by ubiquitin carboxyl-terminal hydrolase L1. J Neurochem 2012; 120:1129-38. [PMID: 22212137 DOI: 10.1111/j.1471-4159.2011.07644.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Deposition of amyloid β protein (Aβ) in the brain is the hallmark of Alzheimer's disease (AD) pathogenesis. Beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) is the β-secretase in vivo essential for generation of Aβ. Previously we demonstrated that BACE1 is ubiquitinated and the degradation of BACE1 is mediated by the ubiquitin-proteasome pathway (UPP). However the mechanism underlying regulation of BACE1 degradation by UPP remains elusive. Ubiquitin carboxyl-terminal hydrolase L1 (UCHL1) is a deubiquitinating enzyme highly specific to neuron, catalyzing the hydrolysis of ubiquitin conjugates from ubiquitinated substrates. UCHL1 regulates ubiquitin-dependent protein degradation. However, whether UCHL1 is particularly involved in the proteasomal degradation of BACE1 and what is the role of UCHL1 in AD pathogenesis remain elusive. To investigate the effect of UCHL1 on BACE1 degradation, HUCH cells, a UCHL1 stably over-expressed HEK293 cell line, was established. We found that inhibition of UCHL1 significantly increased BACE1 protein level in a time-dependent manner. Half life of BACE1 was reduced in HUCH cells compared with HEK. Over-expression of UCHL1 decreased APP C-terminal fragment C99 and Aβ levels in HUCH cells. Moreover, disruption of Uchl1 gene significantly elevated levels of endogenous BACE1, C99 and Aβ in the Uchl1-null gad mice. These results demonstrated that UCHL1 accelerates BACE1 degradation and affects APP processing and Aβ production. This study suggests that potentiation of UCHL1 might be able to reduce the level of BACE1 and Aβ in brain, which makes it a novel target for AD drug development.
Collapse
Affiliation(s)
- Mingming Zhang
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | | | |
Collapse
|
366
|
Mattsson N, Rajendran L, Zetterberg H, Gustavsson M, Andreasson U, Olsson M, Brinkmalm G, Lundkvist J, Jacobson LH, Perrot L, Neumann U, Borghys H, Mercken M, Dhuyvetter D, Jeppsson F, Blennow K, Portelius E. BACE1 inhibition induces a specific cerebrospinal fluid β-amyloid pattern that identifies drug effects in the central nervous system. PLoS One 2012; 7:e31084. [PMID: 22328928 PMCID: PMC3273469 DOI: 10.1371/journal.pone.0031084] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/01/2012] [Indexed: 12/17/2022] Open
Abstract
BACE1 is a key enzyme for amyloid-β (Aβ) production, and an attractive therapeutic target in Alzheimer's disease (AD). Here we report that BACE1 inhibitors have distinct effects on neuronal Aβ metabolism, inducing a unique pattern of secreted Aβ peptides, analyzed in cell media from amyloid precursor protein (APP) transfected cells and in cerebrospinal fluid (CSF) from dogs by immunoprecipitation-mass spectrometry, using several different BACE1 inhibitors. Besides the expected reductions in Aβ1-40 and Aβ1-42, treatment also changed the relative levels of several other Aβ isoforms. In particular Aβ1-34 decreased, while Aβ5-40 increased, and these changes were more sensitive to BACE1 inhibition than the changes in Aβ1-40 and Aβ1-42. The effects on Aβ5-40 indicate the presence of a BACE1 independent pathway of APP degradation. The described CSF Aβ pattern may be used as a pharmacodynamic fingerprint to detect biochemical effects of BACE1-therapies in clinical trials, which might accelerate development of novel therapies.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, Division of Psychiatry Research, University of Zurich, Zurich, Switzerland
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Mikael Gustavsson
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Maria Olsson
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Johan Lundkvist
- Innovative Medicines, Central Nervous System and Pain iMed, Department of Neuroscience, AstraZeneca R&D, Södertälje, Sweden
| | - Laura H. Jacobson
- Neuroscience Discovery, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ludovic Perrot
- Neuroscience Discovery, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ulf Neumann
- Neuroscience Discovery, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Herman Borghys
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Marc Mercken
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Deborah Dhuyvetter
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Fredrik Jeppsson
- Innovative Medicines, Central Nervous System and Pain iMed, Department of Neuroscience, AstraZeneca R&D, Södertälje, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
367
|
Mouton-Liger F, Paquet C, Dumurgier J, Bouras C, Pradier L, Gray F, Hugon J. Oxidative stress increases BACE1 protein levels through activation of the PKR-eIF2α pathway. Biochim Biophys Acta Mol Basis Dis 2012; 1822:885-96. [PMID: 22306812 DOI: 10.1016/j.bbadis.2012.01.009] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 01/18/2012] [Accepted: 01/18/2012] [Indexed: 12/14/2022]
Abstract
Beta-site APP cleaving enzyme 1 (BACE1) is the rate limiting enzyme for accumulation of amyloid β (Aβ)-peptide in the brain in Alzheimer's disease (AD). Oxidative stress (OS) that leads to metabolic dysfunction and apoptosis of neurons in AD enhances BACE1 expression and activity. The activation of c-jun N-terminal kinase (JNK) pathway was proposed to explain the BACE1 mRNA increase under OS. However, little is known about the translational control of BACE1 in OS. Recently, a post-transcriptional increase of BACE1 level controlled by phosphorylation of eIF2α (eukaryotic translation initiation factor-2α) have been described after energy deprivation. PKR (double-stranded RNA dependant protein kinase) is a pro-apoptotic kinase that phosphorylates eIF2α and modulates JNK activation in various cellular stresses. We investigated the relations between PKR, eIF2α and BACE1 in AD brains in APP/PS1 knock-in mice and in hydrogen peroxide-induced OS in human neuroblastoma (SH-SY5Y) cell cultures. Immunoblotting results showed that activated PKR (pPKR) and activated eIF2α (peIF2α) and BACE1 levels are increased in AD cortices and BACE1 correlate with phosphorylated eIF2α levels. BACE1 protein levels are increased in response to OS in SH-SY5Y cells and specific inhibitions of PKR-eIF2α attenuate BACE1 protein levels in this model. Our findings provide a new translational regulation of BACE1, under the control of PKR in OS, where eIF2α phosphorylation regulates BACE1 protein expression.
Collapse
Affiliation(s)
- François Mouton-Liger
- Service d'Histologie et de Biologie du Vieillissement, APHP, Groupe Hospitalier Lariboisière Fernand-Widal Saint-Louis, Université Paris VII, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
368
|
Ding Y, Ko MH, Pehar M, Kotch F, Peters NR, Luo Y, Salamat SM, Puglielli L. Biochemical inhibition of the acetyltransferases ATase1 and ATase2 reduces β-secretase (BACE1) levels and Aβ generation. J Biol Chem 2012; 287:8424-33. [PMID: 22267734 PMCID: PMC3318698 DOI: 10.1074/jbc.m111.310136] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The cellular levels of β-site APP cleaving enzyme 1 (BACE1), the rate-limiting enzyme for the generation of the Alzheimer disease (AD) amyloid β-peptide (Aβ), are tightly regulated by two ER-based acetyl-CoA:lysine acetyltransferases, ATase1 and ATase2. Here we report that both acetyltransferases are expressed in neurons and glial cells, and are up-regulated in the brain of AD patients. We also report the identification of first and second generation compounds that inhibit ATase1/ATase2 and down-regulate the expression levels as well as activity of BACE1. The mechanism of action involves competitive and non-competitive inhibition as well as generation of unstable intermediates of the ATases that undergo degradation.
Collapse
Affiliation(s)
- Yun Ding
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | | | | | | | | | | | | | | |
Collapse
|
369
|
Hook G, Hook V, Kindy M. The cysteine protease inhibitor, E64d, reduces brain amyloid-β and improves memory deficits in Alzheimer's disease animal models by inhibiting cathepsin B, but not BACE1, β-secretase activity. J Alzheimers Dis 2012; 26:387-408. [PMID: 21613740 DOI: 10.3233/jad-2011-110101] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The cysteine protease cathepsin B is a potential drug target for reducing brain amyloid-β (Aβ) and improving memory in Alzheimer's disease (AD), as reduction of cathepsin B in transgenic mice expressing human wild-type amyloid-β protein precursor (AβPP) results in significantly decreased brain Aβ. Cathepsin B cleaves the wild-type β-secretase site sequence in AβPP to produce Aβ, and cathepsin B inhibitors administered to animal models expressing AβPP containing the wild-type β-secretase site sequence reduce brain Aβ in a manner consistent with β-secretase inhibition. But such inhibitors could act either by direct inhibition of cathepsin B β-secretase activity or by off-target inhibition of the other β-secretase, the aspartyl protease BACE1. To evaluate that issue, we orally administered a cysteine protease inhibitor, E64d, to normal guinea pigs or transgenic mice expressing human AβPP, both of which express the human wild-type β-secretase site sequence. In guinea pigs, oral E64d administration caused a dose-dependent reduction of up to 92% in brain, CSF, and plasma of Aβ40 and Aβ42, a reduction of up to 50% in the C-terminal β-secretase fragment (CTFβ), and a 91% reduction in brain cathepsin B activity, but increased brain BACE1 activity by 20%. In transgenic AD mice, oral E64d administration improved memory deficits and reduced brain Aβ40 and Aβ42, amyloid plaque, brain CTFβ, and brain cathepsin B activity, but increased brain BACE1 activity. We conclude that E64d likely reduces brain Aβ by inhibiting cathepsin B and not BACE1 β-secretase activity and that E64d therefore may have potential for treating AD patients.
Collapse
Affiliation(s)
- Gregory Hook
- American Life Science Pharmaceuticals, San Diego, CA 92109, USA.
| | | | | |
Collapse
|
370
|
Sadleir KR, Vassar R. Cdk5 protein inhibition and Aβ42 increase BACE1 protein level in primary neurons by a post-transcriptional mechanism: implications of CDK5 as a therapeutic target for Alzheimer disease. J Biol Chem 2012; 287:7224-35. [PMID: 22223639 DOI: 10.1074/jbc.m111.333914] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The β-secretase enzyme BACE1 initiates production of the amyloid-β (Aβ) peptide that comprises plaques in Alzheimer disease (AD) brain. BACE1 levels are increased in AD, potentially accelerating Aβ generation, but the mechanisms of BACE1 elevation are not fully understood. Cdk5/p25 has been implicated in neurodegeneration and BACE1 regulation, suggesting therapeutic Cdk5 inhibition for AD. In addition, caspase 3 has been implicated in BACE1 elevation. Here, we show that the Cdk5 level and p25:p35 ratio were elevated and correlated with BACE1 level in brains of AD patients and 5XFAD transgenic mice. Mouse primary cortical neurons treated with Aβ42 oligomers had increased BACE1 level and p25:p35 ratio. Surprisingly, the Aβ42-induced BACE1 elevation was not blocked by Cdk5 inhibitors CP68130 and roscovitine, and instead the BACE1 level was increased greater than with Aβ42 treatment alone. Moreover, Cdk5 inhibitors alone elevated BACE1 in a time- and dose-dependent manner that coincided with increased caspase 3 cleavage and decreased Cdk5 level. Caspase 3 inhibitor benzyloxycarbonyl-VAD failed to prevent the Aβ42-induced BACE1 increase. Further experiments suggested that the Aβ42-induced BACE1 elevation was the result of a post-transcriptional mechanism. We conclude that Aβ42 may increase the BACE1 level independently of either Cdk5 or caspase 3 and that Cdk5 inhibition for AD may cause BACE1 elevation, a potentially negative therapeutic outcome.
Collapse
Affiliation(s)
- Katherine R Sadleir
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | |
Collapse
|
371
|
Kumar S, Okello EJ, Harris JR. Experimental inhibition of fibrillogenesis and neurotoxicity by amyloid-beta (Aβ) and other disease-related peptides/proteins by plant extracts and herbal compounds. Subcell Biochem 2012; 65:295-326. [PMID: 23225009 DOI: 10.1007/978-94-007-5416-4_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Amyloid-β (Aβ) fibrillogenesis and associated cyto/neurotoxicity are major pathological events and hallmarks in diseases such as Alzheimer's disease (AD). The understanding of Aβ molecular pathogenesis is currently a pharmacological target for rational drug design and discovery based on reduction of Aβ generation, inhibition of Aβ fibrillogenesis and aggregation, enhancement of Aβ clearance and amelioration of associated cytotoxicity. Molecular mechanisms for other amyloidoses, such as transthyretin amyloidosis, AL-amyloidosis, as well as α-synuclein and prion protein are also pharmacological targets for current drug therapy, design and discovery. We report on natural herbal compounds and extracts that are capable binding to and inhibiting different targets associated with AD and other amyloid-associated diseases, providing a basis for future therapeutic strategies. Many herbal compounds, including curcumin, galantamine, quercetin and other polyphenols, are under active investigation and hold considerable potential for future prophylactic and therapeutic treatment against AD and other neurodegenerative diseases, as well as systemic amyloid diseases. A common emerging theme throughout many studies is the anti-oxidant and anti-inflammatory properties of the compounds or herbal extracts under investigation, within the context of the inhibition of cyto/neurotoxicity and anti-amyloid activity.
Collapse
Affiliation(s)
- Suresh Kumar
- University School of Biotechnology, GGS Indraprastha University, Sector 16C, 10075, Dwarka, Delhi, India,
| | | | | |
Collapse
|
372
|
Meakin P, Harper A, Hamilton D, Gallagher J, McNeilly A, Burgess L, Vaanholt L, Bannon K, Latcham J, Hussain I, Speakman J, Howlett D, Ashford M. Reduction in BACE1 decreases body weight, protects against diet-induced obesity and enhances insulin sensitivity in mice. Biochem J 2012; 441:285-96. [PMID: 21880018 PMCID: PMC3242510 DOI: 10.1042/bj20110512] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 08/18/2011] [Accepted: 08/31/2011] [Indexed: 01/10/2023]
Abstract
Insulin resistance and impaired glucose homoeostasis are important indicators of Type 2 diabetes and are early risk factors of AD (Alzheimer's disease). An essential feature of AD pathology is the presence of BACE1 (β-site amyloid precursor protein-cleaving enzyme 1), which regulates production of toxic amyloid peptides. However, whether BACE1 also plays a role in glucose homoeostasis is presently unknown. We have used transgenic mice to analyse the effects of loss of BACE1 on body weight, and lipid and glucose homoeostasis. BACE1-/- mice are lean, with decreased adiposity, higher energy expenditure, and improved glucose disposal and peripheral insulin sensitivity than wild-type littermates. BACE1-/- mice are also protected from diet-induced obesity. BACE1-deficient skeletal muscle and liver exhibit improved insulin sensitivity. In a skeletal muscle cell line, BACE1 inhibition increased glucose uptake and enhanced insulin sensitivity. The loss of BACE1 is associated with increased levels of UCP1 (uncoupling protein 1) in BAT (brown adipose tissue) and UCP2 and UCP3 mRNA in skeletal muscle, indicative of increased uncoupled respiration and metabolic inefficiency. Thus BACE1 levels may play a critical role in glucose and lipid homoeostasis in conditions of chronic nutrient excess. Therefore strategies that ameliorate BACE1 activity may be important novel approaches for the treatment of diabetes.
Collapse
Key Words
- β-site amyloid precursor protein-cleaving enzyme 1 (bace1)
- glucose uptake
- insulin sensitivity
- liver
- skeletal muscle
- uncoupling protein (ucp)
- aβ, β-amyloid peptide
- ad, alzheimer's disease
- addl, aβ-derived diffusible ligands
- ampk, amp-activated protein kinase
- app, amyloid precursor protein
- bace1, β-site amyloid precursor protein-cleaving enzyme 1
- bat, brown adipose tissue
- dmem, dulbecco's modified eagle's medium
- fbs, fetal bovine serum
- ffa, free fatty acid
- hbs, hepes-buffered saline
- hfd, high-fat diet
- igtt, intraperitoneal glucose tolerance test
- itt, insulin tolerance test
- irs, insulin receptor substrate
- ogtt, oral glucose tolerance test
- pdk, phosphoinositide-dependent kinase
- pkb, protein kinase b
- qmr, quantitative magnetic resonance
- qrt-pcr, quantitative real-time pcr
- rmr, resting metabolic rate
- rq, respiratory quotient
- t4, thyroxine
- tg, triacylglycerol
- ucp, uncoupling protein
- wat, white adipose tissue
- wt, wild-type
Collapse
Affiliation(s)
- Paul J. Meakin
- *Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, U.K
| | - Alex J. Harper
- †Neuroscience Centre of Excellence for Drug Discovery, GlaxoSmithKline R&D, New Frontiers Science Park, Harlow CM19 5AW, U.K
| | - D. Lee Hamilton
- *Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, U.K
| | - Jennifer Gallagher
- *Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, U.K
| | - Alison D. McNeilly
- ‡Division of Neuroscience, University of Dundee, Medical Research Institute, Dundee DD1 9SY, Scotland, U.K
| | - Laura A. Burgess
- *Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, U.K
| | - Lobke M. Vaanholt
- §Aberdeen Centre for Energy Regulation and Obesity, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen AB24 2TZ, Scotland, U.K
| | - Kirsten A. Bannon
- *Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, U.K
| | - Judy Latcham
- †Neuroscience Centre of Excellence for Drug Discovery, GlaxoSmithKline R&D, New Frontiers Science Park, Harlow CM19 5AW, U.K
| | - Ishrut Hussain
- †Neuroscience Centre of Excellence for Drug Discovery, GlaxoSmithKline R&D, New Frontiers Science Park, Harlow CM19 5AW, U.K
| | - John R. Speakman
- §Aberdeen Centre for Energy Regulation and Obesity, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen AB24 2TZ, Scotland, U.K
| | - David R. Howlett
- †Neuroscience Centre of Excellence for Drug Discovery, GlaxoSmithKline R&D, New Frontiers Science Park, Harlow CM19 5AW, U.K
| | - Michael L.J. Ashford
- *Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, U.K
| |
Collapse
|
373
|
Savonenko AV, Melnikova T, Hiatt A, Li T, Worley PF, Troncoso JC, Wong PC, Price DL. Alzheimer's therapeutics: translation of preclinical science to clinical drug development. Neuropsychopharmacology 2012; 37:261-77. [PMID: 21937983 PMCID: PMC3238084 DOI: 10.1038/npp.2011.211] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 08/16/2011] [Accepted: 08/16/2011] [Indexed: 12/15/2022]
Abstract
Over the past three decades, significant progress has been made in understanding the neurobiology of Alzheimer's disease. In recent years, the first attempts to implement novel mechanism-based treatments brought rather disappointing results, with low, if any, drug efficacy and significant side effects. A discrepancy between our expectations based on preclinical models and the results of clinical trials calls for a revision of our theoretical views and questions every stage of translation-from how we model the disease to how we run clinical trials. In the following sections, we will use some specific examples of the therapeutics from acetylcholinesterase inhibitors to recent anti-Aβ immunization and γ-secretase inhibition to discuss whether preclinical studies could predict the limitations in efficacy and side effects that we were so disappointed to observe in recent clinical trials. We discuss ways to improve both the predictive validity of mouse models and the translation of knowledge between preclinical and clinical stages of drug development.
Collapse
Affiliation(s)
- Alena V Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | |
Collapse
|
374
|
Rajapaksha TW, Eimer WA, Bozza TC, Vassar R. The Alzheimer's β-secretase enzyme BACE1 is required for accurate axon guidance of olfactory sensory neurons and normal glomerulus formation in the olfactory bulb. Mol Neurodegener 2011; 6:88. [PMID: 22204380 PMCID: PMC3269394 DOI: 10.1186/1750-1326-6-88] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 12/28/2011] [Indexed: 11/10/2022] Open
Abstract
Background The β-secretase, β-site amyloid precursor protein cleaving enzyme 1 (BACE1), is a prime therapeutic target for lowering cerebral β-amyloid (Aβ) levels in Alzheimer's disease (AD). Clinical development of BACE1 inhibitors is being intensely pursued. However, little is known about the physiological functions of BACE1, and the possibility exists that BACE1 inhibition may cause mechanism-based side effects. Indeed, BACE1-/- mice exhibit a complex neurological phenotype. Interestingly, BACE1 co-localizes with presynaptic neuronal markers, indicating a role in axons and/or terminals. Moreover, recent studies suggest axon guidance molecules are potential BACE1 substrates. Here, we used a genetic approach to investigate the function of BACE1 in axon guidance of olfactory sensory neurons (OSNs), a well-studied model of axon targeting in vivo. Results We bred BACE1-/- mice with gene-targeted mice in which GFP is expressed from the loci of two odorant-receptors (ORs), MOR23 and M72, and olfactory marker protein (OMP) to produce offspring that were heterozygous for MOR23-GFP, M72-GFP, or OMP-GFP and were either BACE1+/+ or BACE1-/-. BACE1-/- mice had olfactory bulbs (OBs) that were smaller and weighed less than OBs of BACE1+/+ mice. In wild-type mice, BACE1 was present in OSN axon terminals in OB glomeruli. In whole-mount preparations and tissue sections, many OB glomeruli from OMP-GFP; BACE1-/- mice were malformed compared to wild-type glomeruli. MOR23-GFP; BACE1-/- mice had an irregular MOR23 glomerulus that was innervated by randomly oriented, poorly fasciculated OSN axons compared to BACE1+/+ mice. Most importantly, M72-GFP; BACE1-/- mice exhibited M72 OSN axons that were mis-targeted to ectopic glomeruli, indicating impaired axon guidance in BACE1-/- mice. Conclusions Our results demonstrate that BACE1 is required for the accurate targeting of OSN axons and the proper formation of glomeruli in the OB, suggesting a role for BACE1 in axon guidance. OSNs continually undergo regeneration and hence require ongoing axon guidance. Neurogenesis and the regeneration of neurons and axons occur in other adult populations of peripheral and central neurons that also require axon guidance throughout life. Therefore, BACE1 inhibitors under development for the treatment of AD may potentially cause axon targeting defects in these neuronal populations as well.
Collapse
Affiliation(s)
- Tharinda W Rajapaksha
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
375
|
Guillot-Sestier MV, Sunyach C, Ferreira ST, Marzolo MP, Bauer C, Thevenet A, Checler F. α-Secretase-derived fragment of cellular prion, N1, protects against monomeric and oligomeric amyloid β (Aβ)-associated cell death. J Biol Chem 2011; 287:5021-32. [PMID: 22184125 DOI: 10.1074/jbc.m111.323626] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In physiological conditions, both β-amyloid precursor protein (βAPP) and cellular prion (PrP(c)) undergo similar disintegrin-mediated α-secretase cleavage yielding N-terminal secreted products referred to as soluble amyloid precursor protein-α (sAPPα) and N1, respectively. We recently demonstrated that N1 displays neuroprotective properties by reducing p53-dependent cell death both in vitro and in vivo. In this study, we examined the potential of N1 as a neuroprotector against amyloid β (Aβ)-mediated toxicity. We first show that both recombinant sAPPα and N1, but not its inactive parent fragment N2, reduce staurosporine-stimulated caspase-3 activation and TUNEL-positive cell death by lowering p53 promoter transactivation and activity in human cells. We demonstrate that N1 also lowers toxicity, cell death, and p53 pathway exacerbation triggered by Swedish mutated βAPP overexpression in human cells. We designed a CHO cell line overexpressing the London mutated βAPP (APP(LDN)) that yields Aβ oligomers. N1 protected primary cultured neurons against toxicity and cell death triggered by oligomer-enriched APP(LDN)-derived conditioned medium. Finally, we establish that N1 also protects neurons against oligomers extracted from Alzheimer disease-affected brain tissues. Overall, our data indicate that a cellular prion catabolite could interfere with Aβ-associated toxicity and that its production could be seen as a cellular protective mechanism aimed at compensating for an sAPPα deficit taking place at the early asymptomatic phase of Alzheimer disease.
Collapse
Affiliation(s)
- Marie-Victoire Guillot-Sestier
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097 CNRS/Université de Nice-Sophia-Antipolis (UNSA), 660 route des Lucioles, Sophia-Antipolis, 06560 Valbonne, France
| | | | | | | | | | | | | |
Collapse
|
376
|
Atwal JK, Chen Y, Chiu C, Mortensen DL, Meilandt WJ, Liu Y, Heise CE, Hoyte K, Luk W, Lu Y, Peng K, Wu P, Rouge L, Zhang Y, Lazarus RA, Scearce-Levie K, Wang W, Wu Y, Tessier-Lavigne M, Watts RJ. A therapeutic antibody targeting BACE1 inhibits amyloid-β production in vivo. Sci Transl Med 2011; 3:84ra43. [PMID: 21613622 DOI: 10.1126/scitranslmed.3002254] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Reducing production of amyloid-β (Aβ) peptide by direct inhibition of the enzymes that process amyloid precursor protein (APP) is a central therapeutic strategy for treating Alzheimer's disease. However, small-molecule inhibitors of the β-secretase (BACE1) and γ-secretase APP processing enzymes have shown a lack of target selectivity and poor penetrance of the blood-brain barrier (BBB). Here, we have developed a high-affinity, phage-derived human antibody that targets BACE1 (anti-BACE1) and is anti-amyloidogenic. Anti-BACE1 reduces endogenous BACE1 activity and Aβ production in human cell lines expressing APP and in cultured primary neurons. Anti-BACE1 is highly selective and does not inhibit the related enzymes BACE2 or cathepsin D. Competitive binding assays and x-ray crystallography indicate that anti-BACE1 binds noncompetitively to an exosite on BACE1 and not to the catalytic site. Systemic dosing of mice and nonhuman primates with anti-BACE1 resulted in sustained reductions in peripheral Aβ peptide concentrations. Anti-BACE1 also reduces central nervous system Aβ concentrations in mouse and monkey, consistent with a measurable uptake of antibody across the BBB. Thus, BACE1 can be targeted in a highly selective manner through passive immunization with anti-BACE1, providing a potential approach for treating Alzheimer's disease. Nevertheless, therapeutic success with anti-BACE1 will depend on improving antibody uptake into the brain.
Collapse
Affiliation(s)
- Jasvinder K Atwal
- Neurodegeneration Labs, Department of Neuroscience, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
377
|
Xu Y, Li MJ, Greenblatt H, Chen W, Paz A, Dym O, Peleg Y, Chen T, Shen X, He J, Jiang H, Silman I, Sussman JL. Flexibility of the flap in the active site of BACE1 as revealed by crystal structures and molecular dynamics simulations. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2011; 68:13-25. [DOI: 10.1107/s0907444911047251] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 11/08/2011] [Indexed: 11/10/2022]
|
378
|
Lu Y, Zhang L, Nolan CE, Becker SL, Atchison K, Robshaw AE, Pustilnik LR, Osgood SM, Miller EH, Stepan AF, Subramanyam C, Efremov I, Hallgren AJ, Riddell D. Quantitative pharmacokinetic/pharmacodynamic analyses suggest that the 129/SVE mouse is a suitable preclinical pharmacology model for identifying small-molecule γ-secretase inhibitors. J Pharmacol Exp Ther 2011; 339:922-34. [PMID: 21930801 DOI: 10.1124/jpet.111.186791] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) poses a serious public health threat to the United States. Disease-modifying drugs slowing AD progression are in urgent need, but they are still unavailable. According to the amyloid cascade hypothesis, inhibition of β- or γ-secretase, key enzymes for the production of amyloid β (Aβ), may be viable mechanisms for the treatment of AD. For the discovery of γ-secretase inhibitors (GSIs), the APP-overexpressing Tg2576 mouse has been the preclinical model of choice, in part because of the ease of detection of Aβ species in its brain, plasma, and cerebrospinal fluid (CSF). Some biological observations and practical considerations, however, argue against the use of the Tg2576 mouse. We reasoned that an animal model would be suitable for GSI discovery if the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a compound for Aβ lowering in this model is predictive of that in human. In this study, we assessed whether the background 129/SVE strain is a suitable preclinical pharmacology model for identifying new GSIs by evaluating the translatability of the intrinsic PK/PD relationships for brain and CSF Aβ across the Tg2576 and 129/SVE mouse and human. Using semimechanistically based PK/PD modeling, our analyses indicated that the intrinsic PK/PD relationship for brain Aβx-42 and CSF Aβx-40 in the 129/SVE mouse is indicative of that for human CSF Aβ. This result, in conjunction with practical considerations, strongly suggests that the 129/SVE mouse is a suitable model for GSI discovery. Concurrently, the necessity and utilities of PK/PD modeling for rational interpretation of Aβ data are established.
Collapse
Affiliation(s)
- Yasong Lu
- Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, CT 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
379
|
Mani V, Ramasamy K, Ahmad A, Parle M, Shah SAA, Majeed ABA. Protective effects of total alkaloidal extract from Murraya koenigii leaves on experimentally induced dementia. Food Chem Toxicol 2011; 50:1036-44. [PMID: 22142688 DOI: 10.1016/j.fct.2011.11.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 11/15/2011] [Accepted: 11/22/2011] [Indexed: 01/26/2023]
Abstract
Dementia is a syndrome of gradual onset and continuous decline of higher cognitive functioning. It is a common disorder in older persons and has become more prevalent today. The fresh leaves of Murraya koenigii are often added to various dishes in Asian countries due to the delicious taste and flavor that they impart. These leaves have also been proven to have health benefits. In the present study, the effect of total alkaloidal extract from M. koenigii leaves (MKA) on cognitive functions and brain cholinesterase activity in mice were determined. In vitro β-secretase 1 (BACE1) inhibitory activity was also evaluated. The total alkaloidal extract was administered orally in three doses (10, 20 and 30 mg/kg) for 15 days to different groups of young and aged mice. Elevated plus maze and passive avoidance apparatus served as the exteroceptive behavioral models for testing memory. Diazepam-, scopolamine-, and ageing-induced amnesia served as the interoceptive behavioral models. MKA (20 and 30 mg/kg, p.o.) showed significant improvement in memory scores of young and aged mice. Furthermore, the same doses of MKA reversed the amnesia induced by scopolamine (0.4 mg/kg, i.p.) and diazepam (1 mg/kg, i.p.). Interestingly, the brain cholinesterase activity was also reduced significantly by total alkaloidal extract of M. koenigii leaves. The IC50 value of MKA against BACE1 was 1.7 μg/mL. In conclusion, this study indicates MKA to be a useful remedy in the management of Alzheimer's disease and dementia.
Collapse
Affiliation(s)
- Vasudevan Mani
- Brain Research Laboratory, Faculty of Pharmacy, Puncak Alam Campus, Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor, Malaysia.
| | | | | | | | | | | |
Collapse
|
380
|
Abstract
Our knowledge of the etiology of Alzheimer's disease (AD) has advanced tremendously since the discovery of amyloid beta (Aβ) aggregation in diseased brains. Accumulating evidence suggests that Aβ plays a causative role in AD. The β-secretase enzyme, beta-site APP cleaving enzyme-1 (BACE1), is also implicated in AD pathogenesis, given that BACE1 cleavage of amyloid precursor protein is the initiating step in the formation of Aβ. As a result, BACE1 inhibition has been branded as a potential AD therapy. In this study, we review the identification and basic characteristics of BACE1, as well as the progress in our understanding of BACE1 cell biology, substrates, and phenotypes of BACE1 knockout mice that are informative about the physiological functions of BACE1 beyond amyloid precursor protein cleavage. These data are crucial for predicting potential mechanism-based toxicity that would arise from inhibiting BACE1 for the treatment or prevention of AD.
Collapse
Affiliation(s)
- Patty C Kandalepas
- Northwestern University, Feinberg School of Medicine, Department of Cell and Molecular Biology, Chicago, Illinois, USA
| | - Robert Vassar
- Northwestern University, Feinberg School of Medicine, Department of Cell and Molecular Biology, Chicago, Illinois, USA
| |
Collapse
|
381
|
Roberts BR, Ryan TM, Bush AI, Masters CL, Duce JA. The role of metallobiology and amyloid-β peptides in Alzheimer’s disease. J Neurochem 2011; 120 Suppl 1:149-166. [DOI: 10.1111/j.1471-4159.2011.07500.x] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
382
|
Zhang H, Ma Q, Zhang YW, Xu H. Proteolytic processing of Alzheimer's β-amyloid precursor protein. J Neurochem 2011; 120 Suppl 1:9-21. [PMID: 22122372 DOI: 10.1111/j.1471-4159.2011.07519.x] [Citation(s) in RCA: 248] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
β-Amyloid precursor protein (APP) is a critical factor in the pathogenesis of Alzheimer's disease (AD). APP undergoes post-translational proteolysis/processing to generate the hydrophobic β-amyloid (Aβ) peptides. Deposition of Aβ in the brain, forming oligomeric Aβ and plaques, is identified as one of the key pathological hallmarks of AD. The processing of APP to generate Aβ is executed by β- and γ-secretase and is highly regulated. Aβ toxicity can lead to synaptic dysfunction, neuronal cell death, impaired learning/memory and abnormal behaviors in AD models in vitro and in vivo. Aside from Aβ, proteolytic cleavages of APP can also give rise to the APP intracellular domain, reportedly involved in multiple types of cellular events such as gene transcription and apoptotic cell death. In addition to amyloidogenic processing, APP can also be cleaved by α-secretase to form a soluble or secreted APP ectodomain (sAPP-α) that has been shown to be mostly neuro-protective. In this review, we describe the mechanisms involved in APP metabolism and the likely functions of its various proteolytic products to give a better understanding of the patho/physiological functions of APP.
Collapse
Affiliation(s)
- Han Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, College of Medicine, Xiamen University, Xiamen, Fujian, China.,Neurodegenerative Disease Research Program, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Qilin Ma
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, College of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, College of Medicine, Xiamen University, Xiamen, Fujian, China.,Neurodegenerative Disease Research Program, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| |
Collapse
|
383
|
Ghosh AK, Brindisi M, Tang J. Developing β-secretase inhibitors for treatment of Alzheimer's disease. J Neurochem 2011; 120 Suppl 1:71-83. [PMID: 22122681 DOI: 10.1111/j.1471-4159.2011.07476.x] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
β-Secretase (memapsin 2; BACE-1) is the first protease in the processing of amyloid precursor protein leading to the production of amyloid-β (Aβ) in the brain. It is believed that high levels of brain Aβ are responsible for the pathogenesis of Alzheimer's disease (AD). Therefore, β-secretase is a major therapeutic target for the development of inhibitor drugs. During the past decade, steady progress has been made in the evolution of β-secretase inhibitors toward better drug properties. Recent inhibitors are potent, selective and have been shown to penetrate the blood-brain barrier to inhibit Aβ levels in the brains of experimental animals. Moreover, continuous administration of a β-secretase inhibitor was shown to rescue age-related cognitive decline in transgenic AD mice. A small number of β-secretase inhibitors have also entered early phase clinical trials. These developments offer some optimism for the clinical development of a disease-modifying drug for AD.
Collapse
Affiliation(s)
- Arun K Ghosh
- Departments of Chemistry and Medicinal Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Margherita Brindisi
- Departments of Chemistry and Medicinal Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Jordan Tang
- Protein Studies Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
384
|
Physiological functions of the amyloid precursor protein secretases ADAM10, BACE1, and Presenilin. Exp Brain Res 2011; 217:331-41. [DOI: 10.1007/s00221-011-2952-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 11/07/2011] [Indexed: 12/16/2022]
|
385
|
Li X, Buxbaum JN. Transthyretin and the brain re-visited: is neuronal synthesis of transthyretin protective in Alzheimer's disease? Mol Neurodegener 2011; 6:79. [PMID: 22112803 PMCID: PMC3267701 DOI: 10.1186/1750-1326-6-79] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Accepted: 11/23/2011] [Indexed: 12/14/2022] Open
Abstract
Since the mid-1990's a trickle of publications from scattered independent laboratories have presented data suggesting that the systemic amyloid precursor transthyretin (TTR) could interact with the amyloidogenic β-amyloid (Aβ) peptide of Alzheimer's disease (AD). The notion that one amyloid precursor could actually inhibit amyloid fibril formation by another seemed quite far-fetched. Further it seemed clear that within the CNS, TTR was only produced in choroid plexus epithelial cells, not in neurons. The most enthusiastic of the authors proclaimed that TTR sequestered Aβ in vivo resulting in a lowered TTR level in the cerebrospinal fluid (CSF) of AD patients and that the relationship was salutary. More circumspect investigators merely showed in vitro interaction between the two molecules. A single in vivo study in Caenorhabditis elegans suggested that wild type human TTR could suppress the abnormalities seen when Aβ was expressed in the muscle cells of the worm. Subsequent studies in human Aβ transgenic mice, including those from our laboratory, also suggested that the interaction reduced the Aβ deposition phenotype. We have reviewed the literature analyzing the relationship including recent data examining potential mechanisms that could explain the effect. We have proposed a model which is consistent with most of the published data and current notions of AD pathogenesis and can serve as a hypothesis which can be tested.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Rd,, MEM-230, La Jolla, CA 92037, USA
| | | |
Collapse
|
386
|
Wen L, Tang FL, Hong Y, Luo SW, Wang CL, He W, Shen C, Jung JU, Xiong F, Lee DH, Zhang QG, Brann D, Kim TW, Yan R, Mei L, Xiong WC. VPS35 haploinsufficiency increases Alzheimer's disease neuropathology. ACTA ACUST UNITED AC 2011; 195:765-79. [PMID: 22105352 PMCID: PMC3257571 DOI: 10.1083/jcb.201105109] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The retromer complex component VPS35 prevents activation of the BACE1 and Aβ production and thus plays an essential role in limiting Alzheimer’s disease neuropathology. VPS35, a major component of the retromer complex, is important for endosome-to-Golgi retrieval of membrane proteins. Although implicated in Alzheimer’s disease (AD), how VPS35 regulates AD-associated pathology is unknown. In this paper, we show that hemizygous deletion of Vps35 in the Tg2576 mouse model of AD led to earlier-onset AD-like phenotypes, including cognitive memory deficits, defective long-term potentiation, and impaired postsynaptic glutamatergic neurotransmission in young adult age. These deficits correlated well with an increase of β-amyloid peptide (Aβ) level in the mutant hippocampus. We further demonstrate that VPS35 is predominantly expressed in pyramidal neurons of young adult hippocampus and interacts with BACE1, a protease responsible for Aβ production. Loss of VPS35 function in the mouse hippocampus increased BACE1 activity. Suppression of VPS35 expression in culture decreased BACE1 trans-Golgi localization but enriched it in endosomes. These results demonstrate an essential role for VPS35 in suppression of AD neuropathology and in inhibition of BACE1 activation and Aβ production by promoting BACE1 endosome-to-Golgi retrieval.
Collapse
Affiliation(s)
- Lei Wen
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
387
|
Chronic stress exacerbates tau pathology, neurodegeneration, and cognitive performance through a corticotropin-releasing factor receptor-dependent mechanism in a transgenic mouse model of tauopathy. J Neurosci 2011; 31:14436-49. [PMID: 21976528 DOI: 10.1523/jneurosci.3836-11.2011] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Because overactivation of the hypothalamic-pituitary-adrenal (HPA) axis occurs in Alzheimer's disease (AD), dysregulation of stress neuromediators may play a mechanistic role in the pathophysiology of AD. However, the effects of stress on tau phosphorylation are poorly understood, and the relationship between corticosterone and corticotropin-releasing factor (CRF) on both β-amyloid (Aβ) and tau pathology remain unclear. Therefore, we first established a model of chronic stress, which exacerbates Aβ accumulation in Tg2576 mice and then extended this stress paradigm to a tau transgenic mouse model with the P301S mutation (PS19) that displays tau hyperphosphorylation, insoluble tau inclusions and neurodegeneration. We show for the first time that both Tg2576 and PS19 mice demonstrate a heightened HPA stress profile in the unstressed state. In Tg2576 mice, 1 month of restraint/isolation (RI) stress increased Aβ levels, suppressed microglial activation, and worsened spatial and fear memory compared with nonstressed mice. In PS19 mice, RI stress promoted tau hyperphosphorylation, insoluble tau aggregation, neurodegeneration, and fear-memory impairments. These effects were not mimicked by chronic corticosterone administration but were prevented by pre-stress administration of a CRF receptor type 1 (CRF(1)) antagonist. The role for a CRF(1)-dependent mechanism was further supported by the finding that mice overexpressing CRF had increased hyperphosphorylated tau compared with wild-type littermates. Together, these results implicate HPA dysregulation in AD neuropathogenesis and suggest that prolonged stress may increase Aβ and tau hyperphosphorylation. These studies also implicate CRF in AD pathophysiology and suggest that pharmacological manipulation of this neuropeptide may be a potential therapeutic strategy for AD.
Collapse
|
388
|
May PC, Dean RA, Lowe SL, Martenyi F, Sheehan SM, Boggs LN, Monk SA, Mathes BM, Mergott DJ, Watson BM, Stout SL, Timm DE, Smith LaBell E, Gonzales CR, Nakano M, Jhee SS, Yen M, Ereshefsky L, Lindstrom TD, Calligaro DO, Cocke PJ, Greg Hall D, Friedrich S, Citron M, Audia JE. Robust central reduction of amyloid-β in humans with an orally available, non-peptidic β-secretase inhibitor. J Neurosci 2011; 31:16507-16. [PMID: 22090477 PMCID: PMC6633289 DOI: 10.1523/jneurosci.3647-11.2011] [Citation(s) in RCA: 304] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 09/01/2011] [Accepted: 09/09/2011] [Indexed: 01/02/2023] Open
Abstract
According to the amyloid cascade hypothesis, cerebral deposition of amyloid-β peptide (Aβ) is critical for Alzheimer's disease (AD) pathogenesis. Aβ generation is initiated when β-secretase (BACE1) cleaves the amyloid precursor protein. For more than a decade, BACE1 has been a prime target for designing drugs to prevent or treat AD. However, development of such agents has turned out to be extremely challenging, with major hurdles in cell penetration, oral bioavailability/metabolic clearance, and brain access. Using a fragment-based chemistry strategy, we have generated LY2811376 [(S)-4-(2,4-difluoro-5-pyrimidin-5-yl-phenyl)-4-methyl-5,6-dihydro-4H-[1,3]thiazin-2-ylamine], the first orally available non-peptidic BACE1 inhibitor that produces profound Aβ-lowering effects in animals. The biomarker changes obtained in preclinical animal models translate into man at doses of LY2811376 that were safe and well tolerated in healthy volunteers. Prominent and long-lasting Aβ reductions in lumbar CSF were measured after oral dosing of 30 or 90 mg of LY2811376. This represents the first translation of BACE1-driven biomarker changes in CNS from preclinical animal models to man. Because of toxicology findings identified in longer-term preclinical studies, this compound is no longer progressing in clinical development. However, BACE1 remains a viable target because the adverse effects reported here were recapitulated in LY2811376-treated BACE1 KO mice and thus are unrelated to BACE1 inhibition. The magnitude and duration of central Aβ reduction obtainable with BACE1 inhibition positions this protease as a tractable small-molecule target through which to test the amyloid hypothesis in man.
Collapse
Affiliation(s)
- Patrick C. May
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Robert A. Dean
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Stephen L. Lowe
- Lilly–National University of Singapore Centre for Clinical Pharmacology, Singapore 117597, Singapore
| | - Ferenc Martenyi
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Scott M. Sheehan
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Leonard N. Boggs
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Scott A. Monk
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Brian M. Mathes
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Dustin J. Mergott
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Brian M. Watson
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Stephanie L. Stout
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - David E. Timm
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | | | | | | | - Stanford S. Jhee
- PAREXEL International Early Phase Los Angeles, Glendale, California 91206
| | - Mark Yen
- PAREXEL International Early Phase Los Angeles, Glendale, California 91206
- California Clinical Trials Medical Group, Glendale, California 91206, and
| | - Larry Ereshefsky
- PAREXEL International Early Phase Los Angeles, Glendale, California 91206
- University of Texas Health Science Center, San Antonio, Texas 98284
| | - Terry D. Lindstrom
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - David O. Calligaro
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Patrick J. Cocke
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - D. Greg Hall
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Stuart Friedrich
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - Martin Citron
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| | - James E. Audia
- Lilly Research Laboratories, Eli Lilly & Co., Indianapolis, Indiana 46285
| |
Collapse
|
389
|
Holler CJ, Webb RL, Laux AL, Beckett TL, Niedowicz DM, Ahmed RR, Liu Y, Simmons CR, Dowling ALS, Spinelli A, Khurgel M, Estus S, Head E, Hersh LB, Murphy MP. BACE2 expression increases in human neurodegenerative disease. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:337-50. [PMID: 22074738 DOI: 10.1016/j.ajpath.2011.09.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 08/02/2011] [Accepted: 09/20/2011] [Indexed: 11/25/2022]
Abstract
β-Secretase, the rate-limiting enzymatic activity in the production of the amyloid-β (Aβ) peptide, is a major target of Alzheimer's disease (AD) therapeutics. There are two forms of the enzyme: β-site Aβ precursor protein cleaving enzyme (BACE) 1 and BACE2. Although BACE1 increases in late-stage AD, little is known about BACE2. We conducted a detailed examination of BACE2 in patients with preclinical to late-stage AD, including amnestic mild cognitive impairment, and age-matched controls, cases of frontotemporal dementia, and Down's syndrome. BACE2 protein and enzymatic activity increased as early as preclinical AD and were found in neurons and astrocytes. Although the levels of total BACE2 mRNA were unchanged, the mRNA for BACE2 splice form C (missing exon 7) increased in parallel with BACE2 protein and activity. BACE1 and BACE2 were strongly correlated with each other at all levels, suggesting that their regulatory mechanisms may be largely shared. BACE2 was also elevated in frontotemporal dementia but not in Down's syndrome, even in patients with substantial Aβ deposition. Thus, expression of both forms of β-secretase are linked and may play a combined role in human neurologic disease. A better understanding of the normal functions of BACE1 and BACE2, and how these change in different disease states, is essential for the future development of AD therapeutics.
Collapse
Affiliation(s)
- Christopher J Holler
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536-0230, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
390
|
Zhao J, O'Connor T, Vassar R. The contribution of activated astrocytes to Aβ production: implications for Alzheimer's disease pathogenesis. J Neuroinflammation 2011; 8:150. [PMID: 22047170 PMCID: PMC3216000 DOI: 10.1186/1742-2094-8-150] [Citation(s) in RCA: 289] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/02/2011] [Indexed: 01/01/2023] Open
Abstract
Background β-Amyloid (Aβ) plays a central role in Alzheimer's disease (AD) pathogenesis. Neurons are major sources of Aβ in the brain. However, astrocytes outnumber neurons by at least five-fold. Thus, even a small level of astrocytic Aβ production could make a significant contribution to Aβ burden in AD. Moreover, activated astrocytes may increase Aβ generation. β-Site APP cleaving enzyme 1 (BACE1) cleavage of amyloid precursor protein (APP) initiates Aβ production. Here, we explored whether pro-inflammatory cytokines or Aβ42 would increase astrocytic levels of BACE1, APP, and β-secretase processing, implying a feed-forward mechanism of astrocytic Aβ production. Methods Mouse primary astrocytes were treated with combinations of LPS, TNF-α, IFN-γ, and IL-1β and analyzed by immunoblot and ELISA for endogenous BACE1, APP, and secreted Aβ40 levels. Inhibition of JAK and iNOS signaling in TNF-α+IFN-γ-stimulated astrocytes was also analyzed. In addition, C57BL/6J or Tg2576 mouse astrocytes were treated with oligomeric or fibrillar Aβ42 and analyzed by immunoblot for levels of BACE1, APP, and APPsβsw. Astrocytic BACE1 and APP mRNA levels were measured by TaqMan RT-PCR. Results TNF-α+IFN-γ stimulation significantly increased levels of astrocytic BACE1, APP, and secreted Aβ40. BACE1 and APP elevations were post-transcriptional at early time-points, but became transcriptional with longer TNF-α+IFN-γ treatment. Despite a ~4-fold increase in astrocytic BACE1 protein level following TNF-α+IFN-γ stimulation, BACE1 mRNA level was significantly decreased suggesting a post-transcriptional mechanism. Inhibition of iNOS and JAK did not reduce TNF-α+IFN-γ-stimulated elevation of astrocytic BACE1, APP, and Aβ40, except that JAK inhibition blocked the APP increase. Finally, oligomeric and fibrillar Aβ42 dramatically increased levels of astrocytic BACE1, APP, and APPsβsw through transcriptional mechanisms, at least in part. Conclusions Cytokines including TNF-α+IFN-γ increase levels of endogenous BACE1, APP, and Aβ and stimulate amyloidogenic APP processing in astrocytes. Oligomeric and fibrillar Aβ42 also increase levels of astrocytic BACE1, APP, and β-secretase processing. Together, our results suggest a cytokine- and Aβ42-driven feed-forward mechanism that promotes astrocytic Aβ production. Given that astrocytes greatly outnumber neurons, activated astrocytes may represent significant sources of Aβ during neuroinflammation in AD.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Cell & Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
391
|
Schechter I, Ziv E. Cathepsins S, B and L with aminopeptidases display β-secretase activity associated with the pathogenesis of Alzheimer's disease. Biol Chem 2011; 392:555-69. [PMID: 21585286 DOI: 10.1515/bc.2011.054] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
β-site APP-cleaving enzyme (BACE1) cleaves the wild type (WT) β-site very slowly (k(cat)/K(m): 46.6 m(-1) s(-1)). Therefore we searched for additional β-secretases and identified three cathepsins that split the WT β-site much faster. Human cathepsin S cleaves the WT β-site (k(cat)/K(m): 54 700 m(-1) s(-1)) 1170-fold faster than BACE1 and cathepsins B and L are 440- and 74-fold faster than BACE1, respectively. These cathepsins split two bonds flanking the WT β-site (K-MD-A), where the K-M bond (85%) is cleaved more efficiently than the D-A bond (15%). Cleavage at the major K-M bond yields Aβ (amyloid β-peptide) extended by N-terminal Met that should be removed to generate Aβ initiated by Asp1. The activity of cytosol and microsomal aminopeptidases on relevant peptides revealed rapid removal of N-terminal Met but not N-terminal Asp. Brain aminopeptidases showed similar specificity. Thus, aminopeptidases would convert Aβ extended by Met into regular Aβ (Asp1) found in amyloid plaques. Earlier studies indicate that Aβ is likely produced in the endosome and lysosome system where cathepsins S, B and L are localized and cysteine cathepsin inhibitors reduce the level of Aβ in cells and animals. Taken together, cathepsins S, B and L deserve further evaluation as therapeutic targets to develop disease modifying drugs to treat Alzheimer's disease.
Collapse
|
392
|
Hook V, Funkelstein L, Wegrzyn J, Bark S, Kindy M, Hook G. Cysteine Cathepsins in the secretory vesicle produce active peptides: Cathepsin L generates peptide neurotransmitters and cathepsin B produces beta-amyloid of Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2011; 1824:89-104. [PMID: 21925292 DOI: 10.1016/j.bbapap.2011.08.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 12/01/2022]
Abstract
Recent new findings indicate significant biological roles of cysteine cathepsin proteases in secretory vesicles for production of biologically active peptides. Notably, cathepsin L in secretory vesicles functions as a key protease for proteolytic processing of proneuropeptides (and prohormones) into active neuropeptides that are released to mediate cell-cell communication in the nervous system for neurotransmission. Moreover, cathepsin B in secretory vesicles has been recently identified as a β-secretase for production of neurotoxic β- amyloid (Aβ) peptides that accumulate in Alzheimer's disease (AD), participating as a notable factor in the severe memory loss in AD. These secretory vesicle functions of cathepsins L and B for production of biologically active peptides contrast with the well-known role of cathepsin proteases in lysosomes for the degradation of proteins to result in their inactivation. The unique secretory vesicle proteome indicates proteins of distinct functional categories that provide the intravesicular environment for support of cysteine cathepsin functions. Features of the secretory vesicle protein systems insure optimized intravesicular conditions that support the proteolytic activity of cathepsins. These new findings of recently discovered biological roles of cathepsins L and B indicate their significance in human health and disease. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.
Collapse
Affiliation(s)
- Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Dept. of Neurosciences, Univ. of Calif., San Diego, USA.
| | | | | | | | | | | |
Collapse
|
393
|
Maltsev AV, Bystryak S, Galzitskaya OV. The role of β-amyloid peptide in neurodegenerative diseases. Ageing Res Rev 2011; 10:440-52. [PMID: 21406255 DOI: 10.1016/j.arr.2011.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 03/03/2011] [Accepted: 03/07/2011] [Indexed: 12/27/2022]
Abstract
Studies of neurodegenerative disorders (NDDs) are drawing more attention of researchers worldwide due to the high incidence of Alzheimer's disease (AD). The pathophysiology of such disorders is, in part, characterized by the transition of a wild-type peptide from its native conformation into a very stable pathological isoform. Subsequently, these abnormal proteins form aggregates of amyloid fibrils that continuously increase in size. Changes in the metabolic processes of neurons (e.g. oxidative stress, hyperphosphorylation of the tau protein, and resulting secondary changes in the cell metabolism) ultimately lead to cell death. We hypothesize that extracellular deposition of β-amyloid peptide fibrils and neurofibrillary tangles represents the body's adaptation mechanism, aimed at preservation of autonomic functioning; while the cognitive decline is severe, the rest of the organ systems remain unaffected and continue to function. This hypothesis is supported by the fact that destruction of pathological plaques, fibrils, and tangles and the use of vaccines targeting β-amyloid result in undesirable side effects. To gain a better understanding of the pathophysiology of Alzheimer's disease and to develop novel therapies, continued studies of the sporadic form of disease and the mechanisms triggering conformational changes in β-amyloid peptide fragments are essential. This review is focused on studies investigating the formation of amyloid fibrils and their role in the pathogenesis of neurodegenerative diseases. In addition, we discuss a related disorder--amyloidosis--where formation of fibrils, tangles, and plaques leads to neuronal death which may occur as a result of a failed adaptation process. Further in-depth investigation and comprehensive analysis of alterations in the metabolism of APP, β-amyloid, and tau protein, which have a pathological effect on cell membrane, alter phosphate exchange, and impair other key metabolic functions of the cell long before the characteristic amyloid deposition takes place, is warranted. A better understanding of intraneuronal processes is crucial in identifying specific inhibitors of pathologic neuronal processes and, consequently, will allow for targeted therapy, thus maximizing efficacy of selected therapeutic regimens.
Collapse
Affiliation(s)
- A V Maltsev
- Russian Gerontological Research Clinical Center, Russian Ministry of Health Care, Moscow, Russia.
| | | | | |
Collapse
|
394
|
Duce JA, Bush AI, Adlard PA. Role of amyloid-β–metal interactions in Alzheimer’s disease. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.43] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There is an evolving field of metallobiology that has begun to describe a key role for bioavailable metals (particularly copper, zinc and iron) in the pathogenesis of Alzheimer’s disease (AD). In particular, there is an apparent failure in metal ion homeostasis, potentially caused by a pathological mislocalization of the metals in the brain, which appears to be an obligatory step in both the precipitation and potentiation of the disease. A number of both preclinical and clinical studies have also provided a strong burden of proof that normalizing metal ion homeostasis represents a valid therapeutic target, and may indeed represent the first disease-modifying strategy for AD. The role of metals in the pathophysiology of AD will be discussed in this article.
Collapse
Affiliation(s)
- James A Duce
- The Mental Health Research Institute, 155 Oak Street, Parkville, Victoria 3052, Australia
- Center for Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Ashley I Bush
- The Mental Health Research Institute, 155 Oak Street, Parkville, Victoria 3052, Australia
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | | |
Collapse
|
395
|
Rabe S, Reichwald J, Ammaturo D, de Strooper B, Saftig P, Neumann U, Staufenbiel M. The Swedish APP mutation alters the effect of genetically reduced BACE1 expression on the APP processing. J Neurochem 2011; 119:231-9. [DOI: 10.1111/j.1471-4159.2011.07412.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
396
|
Chambon C, Wegener N, Gravius A, Danysz W. Behavioural and cellular effects of exogenous amyloid-β peptides in rodents. Behav Brain Res 2011; 225:623-41. [PMID: 21884730 DOI: 10.1016/j.bbr.2011.08.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 08/10/2011] [Accepted: 08/16/2011] [Indexed: 12/29/2022]
Abstract
A better understanding of Alzheimer's disease (AD) and the development of disease modifying therapies are some of the biggest challenges of the 21st century. One of the core features of AD are amyloid plaques composed of amyloid-beta (Aβ) peptides. The first hypothesis proposed that cognitive deficits are linked to plaque-development and transgenic mice have been generated to study this link, thereby providing a good model to develop new therapeutic approaches. Since later it was recognised that in AD patients the cognitive deficit is rather correlated to soluble amyloid levels, consequently, a new hypothesis appeared associating the earliest amyloid toxicity to these soluble species. The purpose of this review is to give a summary of behavioural and cellular data obtained after soluble Aβ peptide administration into rodents' brain, thereby showing that this model is a valid tool to investigate AD pathology when no plaques are present. Additionally, this method offers an excellent, efficient model to test compounds which could act at such early stages of the disease.
Collapse
Affiliation(s)
- Caroline Chambon
- In Vivo Pharmacology, Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, D-60318 Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
397
|
Zhi P, Chia PZC, Chia C, Gleeson PA. Intracellular trafficking of the β-secretase and processing of amyloid precursor protein. IUBMB Life 2011; 63:721-9. [PMID: 21834057 DOI: 10.1002/iub.512] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/11/2011] [Indexed: 12/15/2022]
Abstract
The main component of the amyloid plaques found in the brains of those with Alzheimer's disease (AD) is a polymerized form of the β-amyloid peptide (Aβ) and is considered to play a central role in the pathogenesis of this neurodegenerative disorder. Aβ is derived from the proteolytic processing of the amyloid precursor protein (APP). Beta site APP-cleaving enzyme, BACE1 (also known as β-secretase) is a membrane-bound aspartyl protease responsible for the initial step in the generation of Aβ peptide and is thus a prime target for therapeutic intervention. Substantive evidence now indicates that the processing of APP by BACE1 is regulated by the intracellular sorting of the enzyme and, moreover, perturbations in these intracellular trafficking pathways have been linked to late-onset AD. In this review, we highlight the recent advances in the understanding of the regulation of the intracellular sorting of BACE1 and APP and illustrate why the trafficking of these cargos represent a key issue for understanding the membrane-mediated events associated with the generation of the neurotoxic Aβ products in AD.
Collapse
Affiliation(s)
- Pei Zhi
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
398
|
Abstract
Sphingosine kinase (SphK) 1 and 2 phosphorylate sphingosine to generate sphingosine-1-phosphate (S1P), a pluripotent lipophilic mediator implicated in a variety of cellular events. Here we show that the activity of β-site APP cleaving enzyme-1 (BACE1), the rate-limiting enzyme for amyloid-β peptide (Aβ) production, is modulated by S1P in mouse neurons. Treatment by SphK inhibitor, RNA interference knockdown of SphK, or overexpression of S1P degrading enzymes decreased BACE1 activity, which reduced Aβ production. S1P specifically bound to full-length BACE1 and increased its proteolytic activity, suggesting that cellular S1P directly modulates BACE1 activity. Notably, the relative activity of SphK2 was upregulated in the brains of patients with Alzheimer's disease. The unique modulatory effect of cellular S1P on BACE1 activity is a novel potential therapeutic target for Alzheimer's disease.
Collapse
|
399
|
Abstract
Alzheimer's disease ranks the first cause for senile dementia. The amyloid cascade is proposed to contribute to the pathogenesis of this disease. In this cascade, amyloid β peptide (Aβ) is produced through a sequential cleavage of amyloid precursor protein (APP) by β and γ secretases, while its cleavage by α secretase precludes Aβ production and generates neurotrophic sAPPα. Thus, enhancing α secretase activity or suppressing β and γ cleavage may reduce Aβ formation and ameliorate the pathological process of the disease. Several regulatory mechanisms of APP cleavage have been established. The present review mainly summarizes the signaling pathways pertinent to the regulation of APP β cleavage.
Collapse
Affiliation(s)
- Jun-Feng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Shanghai 200031, China
| | | | | |
Collapse
|
400
|
Cheng Y, Judd TC, Bartberger MD, Brown J, Chen K, Fremeau RT, Hickman D, Hitchcock SA, Jordan B, Li V, Lopez P, Louie SW, Luo Y, Michelsen K, Nixey T, Powers TS, Rattan C, Sickmier EA, St Jean DJ, Wahl RC, Wen PH, Wood S. From fragment screening to in vivo efficacy: optimization of a series of 2-aminoquinolines as potent inhibitors of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1). J Med Chem 2011; 54:5836-57. [PMID: 21707077 DOI: 10.1021/jm200544q] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Using fragment-based screening of a focused fragment library, 2-aminoquinoline 1 was identified as an initial hit for BACE1. Further SAR development was supported by X-ray structures of BACE1 cocrystallized with various ligands and molecular modeling studies to expedite the discovery of potent compounds. These strategies enabled us to integrate the C-3 side chain on 2-aminoquinoline 1 extending deep into the P2' binding pocket of BACE1 and enhancing the ligand's potency. We were able to improve the BACE1 potency to subnanomolar range, over 10(6)-fold more potent than the initial hit (900 μM). Further elaboration of the physical properties of the lead compounds to those more consistent with good blood-brain barrier permeability led to inhibitors with greatly improved cellular activity and permeability. Compound 59 showed an IC(50) value of 11 nM on BACE1 and cellular activity of 80 nM. This compound was advanced into rat pharmacokinetic and pharmacodynamic studies and demonstrated significant reduction of Aβ levels in cerebrospinal fluid (CSF).
Collapse
Affiliation(s)
- Yuan Cheng
- Chemistry Research and Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|