201
|
Decourt B, Mobley W, Reiman E, Shah RJ, Sabbagh MN. Recent Perspectives on APP, Secretases, Endosomal Pathways and How they Influence Alzheimer's Related Pathological Changes in Down Syndrome. ACTA ACUST UNITED AC 2013; Suppl 7:002. [PMID: 24782952 PMCID: PMC4000700 DOI: 10.4172/2161-0460.s7-002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Down syndrome is one of the most common genetic conditions occurring in one in 700 live births. The trisomy of chromosome 21 causes over-expression of APP which in turn is indicated in the increased production of Aβ associated with AD. This makes DS the most common presenile form of AD exceeding PS1 and PS2 FAD. Since a majority of DS individuals develop dementia, it is important to examine whether DS and sporadic AD share common features, for example, to anticipate shared treatments in the future. Here we explore commonalities and differences for secretases and endosomal pathways in DS and AD.
Collapse
Affiliation(s)
- Boris Decourt
- Banner Sun Health Research Institute, Sun City AZ, USA
| | | | - Eric Reiman
- Banner Alzheimer's Institute, Phoenix AZ, USA
| | | | | |
Collapse
|
202
|
Ly PTT, Wu Y, Zou H, Wang R, Zhou W, Kinoshita A, Zhang M, Yang Y, Cai F, Woodgett J, Song W. Inhibition of GSK3β-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J Clin Invest 2012. [PMID: 23202730 DOI: 10.1172/jci64516] [Citation(s) in RCA: 340] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Deposition of amyloid β protein (Aβ) to form neuritic plaques in the brain is the pathological hallmark of Alzheimer's disease (AD). Aβ is generated from sequential cleavages of the β-amyloid precursor protein (APP) by the β- and γ-secretases, and β-site APP-cleaving enzyme 1 (BACE1) is the β-secretase essential for Aβ generation. Previous studies have indicated that glycogen synthase kinase 3 (GSK3) may play a role in APP processing by modulating γ-secretase activity, thereby facilitating Aβ production. There are two highly conserved isoforms of GSK3: GSK3α and GSK3β. We now report that specific inhibition of GSK3β, but not GSK3α, reduced BACE1-mediated cleavage of APP and Aβ production by decreasing BACE1 gene transcription and expression. The regulation of BACE1 gene expression by GSK3β was dependent on NF-κB signaling. Inhibition of GSK3 signaling markedly reduced Aβ deposition and neuritic plaque formation, and rescued memory deficits in the double transgenic AD model mice. These data provide evidence for regulation of BACE1 expression and AD pathogenesis by GSK3β and that inhibition of GSK3 signaling can reduce Aβ neuropathology and alleviate memory deficits in AD model mice. Our study suggests that interventions that specifically target the β-isoform of GSK3 may be a safe and effective approach for treating AD.
Collapse
Affiliation(s)
- Philip T T Ly
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Lu Q, Chen WY, Zhu ZY, Chen J, Xu YC, Kaewpet M, Rukachaisirikul V, Chen LL, Shen X. L655,240, acting as a competitive BACE1 inhibitor, efficiently decreases β-amyloid peptide production in HEK293-APPswe cells. Acta Pharmacol Sin 2012; 33:1459-68. [PMID: 22842730 DOI: 10.1038/aps.2012.74] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
AIM To identify a small molecule L655,240 as a novel β-secretase (BACE1) inhibitor and to investigate its effects on β-amyloid (Aβ) generation in vitro. METHODS Fluorescence resonance energy transfer (FRET) was used to characterize the inhibitory effect of L655,240 on BACE1. Surface plasmon resonance (SPR) technology-based assay was performed to study the binding affinity of L655,240 for BACE1. The selectivity of L655,240 toward BACE1 over other aspartic proteases was determined with enzymatic assay. The effects of L655,240 on Aβ40, Aβ42, and sAPPβ production were studied in HEK293 cells stably expressing APP695 Swedish mutant(K595N/M596L) (HEK293-APPswe cells). The activities of BACE1, γ-secretase and α-secretase were assayed, and both the mRNA and protein levels of APP and BACE1 were evaluated using real-time PCR (RT-PCR) and Western blot analysis. RESULTS L655,240 was determined to be a competitive, selective BACE1 inhibitor (IC(50)=4.47±1.37 μmol/L), which bound to BACE1 directly (K(D)=17.9±0.72 μmol/L). L655,240 effectively reduced Aβ40, Aβ42, and sAPPβ production by inhibiting BACE1 without affecting the activities of γ-secretase and α-secretase in HEK293-APPswe cells. L655,240 has no effect on APP and BACE1 mRNA or protein levels in HEK293-APPswe cells. CONCLUSION The small molecule L655,240 is a novel BACE1 inhibitor that can effectively decreases Aβ production in vitro, thereby highlighting its therapeutic potential for the treatment of Alzheimer's disease.
Collapse
|
204
|
Razzaghi-Asl N, Ebadi A, Edraki N, Shahabipour S, Miri R. Ab initio modeling of a potent isophthalamide-based BACE-1 inhibitor: amino acid decomposition analysis. Med Chem Res 2012. [DOI: 10.1007/s00044-012-0277-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
205
|
Holsinger RMD, Goense N, Bohorquez J, Strappe P. Splice variants of the Alzheimer's disease beta-secretase, BACE1. Neurogenetics 2012; 14:1-9. [PMID: 23142975 DOI: 10.1007/s10048-012-0348-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/15/2012] [Indexed: 10/27/2022]
Abstract
Cleavage of the amyloid precursor protein by enzymes commonly referred to as β- and γ-secretase constitute an important process in the pathogenesis of Alzheimer's disease (AD). The regulation of this process is therefore an important subject of investigation. Numerous sources of endogenous regulation have been identified, and one of these is the relative abundance and regulation of splice variants of the β-secretase, BACE1 (β-site amyloid precursor protein cleaving enzyme 1). In this review, we will briefly discuss the main characteristics of BACE1, review the different variants of this enzyme that have been identified to date, and highlight their possible implication in AD.
Collapse
Affiliation(s)
- R M Damian Holsinger
- Laboratory of Molecular Neuroscience, Brain and Mind Research Institute, The University of Sydney, Camperdown, NSW 2050, Australia.
| | | | | | | |
Collapse
|
206
|
Stamford AW, Scott JD, Li SW, Babu S, Tadesse D, Hunter R, Wu Y, Misiaszek J, Cumming JN, Gilbert EJ, Huang C, McKittrick B, Hong L, Guo T, Zhu Z, Strickland C, Orth P, Voigt J, Kennedy ME, Chen X, Kuvelkar R, Hodgson R, Hyde L, Cox K, Favreau L, Parker EM, Greenlee WJ. Discovery of an Orally Available, Brain Penetrant BACE1 Inhibitor that Affords Robust CNS Aβ Reduction. ACS Med Chem Lett 2012; 3:897-902. [PMID: 23412139 PMCID: PMC3568987 DOI: 10.1021/ml3001165] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/12/2012] [Indexed: 01/16/2023] Open
Abstract
Inhibition of BACE1 to prevent brain Aβ peptide formation is a potential disease-modifying approach to the treatment of Alzheimer's disease. Despite over a decade of drug discovery efforts, the identification of brain-penetrant BACE1 inhibitors that substantially lower CNS Aβ levels following systemic administration remains challenging. In this report we describe structure-based optimization of a series of brain-penetrant BACE1 inhibitors derived from an iminopyrimidinone scaffold. Application of structure-based design in tandem with control of physicochemical properties culminated in the discovery of compound 16, which potently reduced cortex and CSF Aβ40 levels when administered orally to rats.
Collapse
Affiliation(s)
- Andrew W. Stamford
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jack D. Scott
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Sarah W. Li
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Suresh Babu
- Ligand Pharmaceuticals, Inc.,
3000 Eastpark Boulevard Cranbury, New Jersey 08512, United States
| | - Dawit Tadesse
- Ligand Pharmaceuticals, Inc.,
3000 Eastpark Boulevard Cranbury, New Jersey 08512, United States
| | - Rachael Hunter
- Ligand Pharmaceuticals, Inc.,
3000 Eastpark Boulevard Cranbury, New Jersey 08512, United States
| | - Yusheng Wu
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jeffrey Misiaszek
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jared N. Cumming
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric J. Gilbert
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Chunli Huang
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brian
A. McKittrick
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Liwu Hong
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Tao Guo
- Ligand Pharmaceuticals, Inc.,
3000 Eastpark Boulevard Cranbury, New Jersey 08512, United States
| | - Zhaoning Zhu
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Corey Strickland
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Peter Orth
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes
H. Voigt
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Matthew E. Kennedy
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xia Chen
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Reshma Kuvelkar
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert Hodgson
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn
A. Hyde
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kathleen Cox
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Leonard Favreau
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - William J. Greenlee
- Departments
of Medicinal Chemistry, Neurobiology, Drug Metabolism, and Global Structural Chemistry, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
207
|
Vincent KJ, Zurini M. Current strategies in antibody engineering: Fc engineering and pH-dependent antigen binding, bispecific antibodies and antibody drug conjugates. Biotechnol J 2012; 7:1444-50. [DOI: 10.1002/biot.201200250] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/13/2012] [Accepted: 10/01/2012] [Indexed: 12/19/2022]
|
208
|
Abstract
Significant insights into the function of genes associated with Alzheimer disease and related dementias have occurred through studying genetically modified animals. Although none of the existing models fully reproduces the complete spectrum of this insidious human disease, critical aspects of Alzheimer pathology and disease processes can be experimentally recapitulated. Genetically modified animal models have helped advance our understanding of the underlying mechanisms of disease and have proven to be invaluable in the preclinical evaluation of potential therapeutic interventions. Continuing refinement and evolution to yield the next generation of animal models will facilitate successes in producing greater translational concordance between preclinical studies and human clinical trials and eventually lead to the introduction of novel therapies into clinical practice.
Collapse
Affiliation(s)
- Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, Department of Neurobiology and Behavior, University of California, Irvine, 92697-4545, USA.
| | | |
Collapse
|
209
|
Kang EL, Biscaro B, Piazza F, Tesco G. BACE1 protein endocytosis and trafficking are differentially regulated by ubiquitination at lysine 501 and the Di-leucine motif in the carboxyl terminus. J Biol Chem 2012; 287:42867-80. [PMID: 23109336 DOI: 10.1074/jbc.m112.407072] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
β-Site amyloid precursor protein-cleaving enzyme (BACE1) is a membrane-tethered member of the aspartyl proteases that has been identified as β-secretase. BACE1 is targeted through the secretory pathway to the plasma membrane and then is internalized to endosomes. Sorting of membrane proteins to the endosomes and lysosomes is regulated by the interaction of signals present in their carboxyl-terminal fragment with specific trafficking molecules. The BACE1 carboxyl-terminal fragment contains a di-leucine sorting signal ((495)DDISLL(500)) and a ubiquitination site at Lys-501. Here, we report that lack of ubiquitination at Lys-501 (BACE1K501R) does not affect the rate of endocytosis but produces BACE1 stabilization and accumulation of BACE1 in early and late endosomes/lysosomes as well as at the cell membrane. In contrast, the disruption of the di-leucine motif (BACE1LLAA) greatly impairs BACE1 endocytosis and produces a delayed retrograde transport of BACE1 to the trans-Golgi network (TGN) and a delayed delivery of BACE1 to the lysosomes, thus decreasing its degradation. Moreover, the combination of the lack of ubiquitination at Lys-501 and the disruption of the di-leucine motif (BACE1LLAA/KR) produces additive effects on BACE1 stabilization and defective internalization. Finally, BACE1LLAA/KR accumulates in the TGN, while its levels are decreased in EEA1-positive compartments indicating that both ubiquitination at Lys-501 and the di-leucine motif are necessary for the trafficking of BACE1 from the TGN to early endosomes. Our studies have elucidated a differential role for the di-leucine motif and ubiquitination at Lys-501 in BACE1 endocytosis, trafficking, and degradation and suggest the involvement of multiple adaptor molecules.
Collapse
Affiliation(s)
- Eugene L Kang
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | | | |
Collapse
|
210
|
Chami L, Checler F. BACE1 is at the crossroad of a toxic vicious cycle involving cellular stress and β-amyloid production in Alzheimer's disease. Mol Neurodegener 2012; 7:52. [PMID: 23039869 PMCID: PMC3507664 DOI: 10.1186/1750-1326-7-52] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/03/2012] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex age-related pathology, the etiology of which has not been firmly delineated. Among various histological stigmata, AD-affected brains display several cellular dysfunctions reflecting enhanced oxidative stress, inflammation process and calcium homeostasis disturbance. Most of these alterations are directly or indirectly linked to amyloid β-peptides (Aβ), the production, molecular nature and biophysical properties of which likely conditions the degenerative process. It is particularly noticeable that, in a reverse control process, the above-described cellular dysfunctions alter Aβ peptides levels. β-secretase βAPP-cleaving enzyme 1 (BACE1) is a key molecular contributor of this cross-talk. This enzyme is responsible for the primary cleavage generating the N-terminus of “full length” Aβ peptides and is also transcriptionally induced by several cellular stresses. This review summarizes data linking brain insults to AD-like pathology and documents the key role of BACE1 at the cross-road of a vicious cycle contributing to Aβ production.
Collapse
Affiliation(s)
- Linda Chami
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 06560 Valbonne, France
| | | |
Collapse
|
211
|
Hitt B, Riordan SM, Kukreja L, Eimer WA, Rajapaksha TW, Vassar R. β-Site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1)-deficient mice exhibit a close homolog of L1 (CHL1) loss-of-function phenotype involving axon guidance defects. J Biol Chem 2012; 287:38408-25. [PMID: 22988240 DOI: 10.1074/jbc.m112.415505] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACE1 is the β-secretase enzyme that initiates production of the β-amyloid peptide involved in Alzheimer disease. However, little is known about the functions of BACE1. BACE1-deficient mice exhibit mild but complex neurological phenotypes suggesting therapeutic BACE1 inhibition may not be completely free of mechanism-based side effects. Recently, we have reported that BACE1 null mice have axon guidance defects in olfactory sensory neuron projections to glomeruli in the olfactory bulb. Here, we show that BACE1 deficiency also causes an axon guidance defect in the hippocampus, a shortened and disorganized infrapyramidal bundle of the mossy fiber projection from the dentate gyrus to CA3. Although we observed that a classical axon guidance molecule, EphA4, was cleaved by BACE1 when co-expressed with BACE1 in HEK293 cells, we could find no evidence of BACE1 processing of EphA4 in the brain. Remarkably, we discovered that the axon guidance defects of BACE1(-/-) mice were strikingly similar to those of mice deficient in a recently identified BACE1 substrate, the neural cell adhesion molecule close homolog of L1 (CHL1) that is involved in neurite outgrowth. CHL1 undergoes BACE1-dependent processing in BACE1(+/+), but not BACE1(-/-), hippocampus, and olfactory bulb, indicating that CHL1 is a BACE1 substrate in vivo. Finally, BACE1 and CHL1 co-localize in the terminals of hippocampal mossy fibers, olfactory sensory neuron axons, and growth cones of primary hippocampal neurons. We conclude that BACE1(-/-) axon guidance defects are likely the result of abrogated BACE1 processing of CHL1 and that BACE1 deficiency produces a CHL1 loss-of-function phenotype. Our results imply the possibility that axon mis-targeting may occur in adult neurogenic and/or regenerating neurons as a result of chronic BACE1 inhibition and add a note of caution to BACE1 inhibitor development.
Collapse
Affiliation(s)
- Brian Hitt
- Department of Cell and Molecular Biology, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
212
|
Swahn BM, Kolmodin K, Karlström S, von Berg S, Söderman P, Holenz J, Berg S, Lindström J, Sundström M, Turek D, Kihlström J, Slivo C, Andersson L, Pyring D, Rotticci D, Öhberg L, Kers A, Bogar K, von Kieseritzky F, Bergh M, Olsson LL, Janson J, Eketjäll S, Georgievska B, Jeppsson F, Fälting J. Design and Synthesis of β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE1) Inhibitors with in Vivo Brain Reduction of β-Amyloid Peptides. J Med Chem 2012; 55:9346-61. [DOI: 10.1021/jm3009025] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lise-Lotte Olsson
- Discovery Sciences, AstraZeneca R&D Mölndal, SE-43183 Mölndal, Sweden
| | | | | | | | | | | |
Collapse
|
213
|
Huang H, La DS, Cheng AC, Whittington DA, Patel VF, Chen K, Dineen TA, Epstein O, Graceffa R, Hickman D, Kiang YH, Louie S, Luo Y, Wahl RC, Wen PH, Wood S, Fremeau RT. Structure- and Property-Based Design of Aminooxazoline Xanthenes as Selective, Orally Efficacious, and CNS Penetrable BACE Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2012; 55:9156-69. [DOI: 10.1021/jm300598e] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hongbing Huang
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Daniel S. La
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Alan C. Cheng
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Douglas A. Whittington
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Vinod F. Patel
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Kui Chen
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Thomas A. Dineen
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Oleg Epstein
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Russell Graceffa
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Dean Hickman
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Y.-H. Kiang
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Steven Louie
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Yi Luo
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Robert C. Wahl
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Paul H. Wen
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Stephen Wood
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| | - Robert T. Fremeau
- Department
of Medicinal Chemistry and ‡Department of Molecular Structure, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts
02142, United States
- Department
of Neuroscience, ∥Department of Pharmacokinetics and Drug Metabolism, ⊥Department of HTS and Molecular
Pharmacology, and #Department of Pharmaceutics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United
States
| |
Collapse
|
214
|
Sund C, Belda O, Borkakoti N, Lindberg J, Derbyshire D, Vrang L, Hamelink E, Åhgren C, Woestenenk E, Wikström K, Eneroth A, Lindström E, Kalayanov G. Design and synthesis of potent hydroxyethylamine (HEA) BACE-1 inhibitors carrying prime side 4,5,6,7-tetrahydrobenzazole and 4,5,6,7-tetrahydropyridinoazole templates. Bioorg Med Chem Lett 2012; 22:6721-7. [PMID: 23010268 DOI: 10.1016/j.bmcl.2012.08.097] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 08/22/2012] [Accepted: 08/24/2012] [Indexed: 01/05/2023]
Abstract
A set of low molecular weight compounds containing a hydroxyethylamine (HEA) core structure with different prime side alkyl substituted 4,5,6,7-tetrahydrobenzazoles and one 4,5,6,7-tetrahydropyridinoazole was synthesized. Striking differences were observed on potencies in the BACE-1 enzymatic and cellular assays depending on the nature of the heteroatoms in the bicyclic ring, from the low active compound 4 to inhibitor 6, displaying BACE-1 IC(50) values of 44 nM (enzyme assay) and 65 nM (cell-based assay).
Collapse
|
215
|
Schenk D, Basi GS, Pangalos MN. Treatment strategies targeting amyloid β-protein. Cold Spring Harb Perspect Med 2012; 2:a006387. [PMID: 22951439 DOI: 10.1101/cshperspect.a006387] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the advent of the key discovery in the mid-1980s that the amyloid β-protein (Aβ) is the core constituent of the amyloid plaque pathology found in Alzheimer disease (AD), an intensive effort has been underway to attempt to mitigate its role in the hope of treating the disease. This effort fully matured when it was clarified that the Aβ is a normal product of cleavage of the amyloid precursor protein, and well-defined proteases for this process were identified. Further therapeutic options have been developed around the concept of anti-Aβ aggregation inhibitors and the surprising finding that immunization with Aβ itself leads to reduction of pathology in animal models of the disease. Here we review the progress in this field toward the goal of targeting Aβ for treatment and prevention of AD and identify some of the major challenges for the future of this area of medicine.
Collapse
Affiliation(s)
- Dale Schenk
- Netotope Biosciences Inc., San Francisco, CA 94080, USA
| | | | | |
Collapse
|
216
|
Ameliorative effects of curculigoside from Curculigo orchioides Gaertn on learning and memory in aged rats. Molecules 2012; 17:10108-18. [PMID: 22922281 PMCID: PMC6268871 DOI: 10.3390/molecules170910108] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/29/2012] [Accepted: 08/09/2012] [Indexed: 11/17/2022] Open
Abstract
This study was designed to evaluate the ameliorating effects of curculigoside from Curculigo orchioides Gaertn on learning and memory in aged rats. In the present study, the ameliorating effects of curculigoside were determined through animal behaviour studies (including step-down test and Y-maze test), and the possible mechanisms were explored by evaluation of the activity of acetylcholinesterase (AchE) and determination of the expression of BACE1. Oral adminstration of the curculigoside (20, 40 mg/kg/day) for 14 days can significantly improve the latency and number of errors in aged rats based on the behaviour study results. In addition, the activity of AchE can be decreased by treatment of the curculigoside (10, 20, 40 mg/kg/day). Moreover, the expression of BACE1 can be down-regulated in the hippocampus of aged rats treated with curculigoside. The results of our present work have indicated that curculigoside can improve cognitive function in aged animals, possibly by decreasing the activity of AchE in the cerebra and inhibiting the expression of BACE1 in the hippocampus. In conclusion, our results suggested that curculigoside can be possible developed as a new drug for the treatment of Alzheimer's disease in the future.
Collapse
|
217
|
Sathya M, Premkumar P, Karthick C, Moorthi P, Jayachandran KS, Anusuyadevi M. BACE1 in Alzheimer's disease. Clin Chim Acta 2012; 414:171-8. [PMID: 22926063 DOI: 10.1016/j.cca.2012.08.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 08/08/2012] [Accepted: 08/15/2012] [Indexed: 12/18/2022]
Abstract
Targeting BACE1 (β-site APP cleaving enzyme 1 or β-secretase) is the focus of Alzheimer's disease (AD) research because this aspartyl protease is involved in the abnormal production of β amyloid plaques (Aβ), the hallmark of its pathophysiology. Evidence suggests that there is a strong connection between AD and BACE1. As such, strategies to inhibit Aβ formation in the brain should prove beneficial for AD treatment. Aβ, the product of the large type1 trans-membrane protein amyloid precursor protein (APP), is produced in a two-step proteolytic process initiated by BACE1 (β-secretase) and followed by γ-secretase. Due to its apparent rate limiting function, BACE1 appears to be a prime target to prevent Aβ generation in AD. Following its discovery, the BACE1 has been cloned, its structure solved, novel physiologic substrates discovered and numerous inhibitors developed. This review focuses on elucidating the role of BACE1 to facilitate drug development in the treatment of AD.
Collapse
Affiliation(s)
- M Sathya
- Department of Biochemistry, Bharathidasan University, Trichy 24, India
| | | | | | | | | | | |
Collapse
|
218
|
Kindy MS, Yu J, Zhu H, El-Amouri SS, Hook V, Hook GR. Deletion of the cathepsin B gene improves memory deficits in a transgenic ALZHeimer's disease mouse model expressing AβPP containing the wild-type β-secretase site sequence. J Alzheimers Dis 2012; 29:827-40. [PMID: 22337825 DOI: 10.3233/jad-2012-111604] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Therapeutic agents that improve the memory loss of Alzheimer's disease (AD) may eventually be developed if drug targets are identified that improve memory deficits in appropriate AD animal models. One such target is β-secretase which, in most AD patients, cleaves the wild-type (WT) β-secretase site sequence of the amyloid-β protein precursor (AβPP) to produce neurotoxic amyloid-β (Aβ). Thus, an animal model representing most AD patients for evaluating β-secretase effects on memory deficits is one that expresses human AβPP containing the WT β-secretase site sequence. BACE1 and cathepsin B (CatB) proteases have β-secretase activity, but gene knockout studies have not yet validated that the absence of these proteases improves memory deficits in such an animal model. This study assessed the effects of deleting these protease genes on memory deficits in the AD mouse model expressing human AβPP containing the WT β-secretase site sequence and the London γ-secretase site (AβPPWT/Lon mice). Knockout of the CatB gene in the AβPPWT/Lon mice improved memory deficits and altered the pattern of Aβ-related biomarkers in a manner consistent with CatB having WT β-secretase activity. But deletion of the BACE1 gene had no effect on these parameters in the AβPPWT/Lon mice. These data are the first to show that knockout of a putative β-secretase gene results in improved memory in an AD animal model expressing the WT β-secretase site sequence of AβPP, present in the majority of AD patients. CatB may be an effective drug target for improving memory deficits in most AD patients.
Collapse
Affiliation(s)
- Mark S Kindy
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | |
Collapse
|
219
|
Koike MA, Lin AJ, Pham J, Nguyen E, Yeh JJ, Rahimian R, Tromberg BJ, Choi B, Green KN, LaFerla FM. APP knockout mice experience acute mortality as the result of ischemia. PLoS One 2012; 7:e42665. [PMID: 22912719 PMCID: PMC3415410 DOI: 10.1371/journal.pone.0042665] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/10/2012] [Indexed: 01/26/2023] Open
Abstract
The incidence of Alzheimer’s disease increases in people who have had an ischemic episode. Furthermore, APP expression is increased following ischemic or hypoxic conditions, as is the production of the Aβ peptide. To address the question of why APP and Aβ are increased in hypoxic and ischemic conditions we induced an ischemic episode in APP knockout mice (APP−/−) and BACE1 knockout mice (BACE−/−). We find that both APP−/− and BACE−/− mice have a dramatically increased risk of mortality as a result of cerebral ischemia. Furthermore, APP knockout mice have reduced cerebral blood flow in response to hypoxia, while wild-type mice maintain or increase cerebral blood flow to the same conditions. The transcription factor, serum response factor (SRF), and calcium-binding molecule, calsequestrin, both involved in vascular regulation, are significantly altered in the brains of APP−/− mice compared to wild type controls. These results show that APP regulates cerebral blood flow in response to hypoxia, and that it, and its cleavage fragments, are crucial for rapid adaptation to ischemic conditions.
Collapse
Affiliation(s)
- Maya A. Koike
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, United States of America
| | - Alexander J. Lin
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, United States of America
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Jonathan Pham
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Elaine Nguyen
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
| | - James J. Yeh
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
| | - Rombod Rahimian
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
| | - Bruce J. Tromberg
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Bernard Choi
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California, United States of America
| | - Kim N. Green
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, United States of America
- * E-mail: (FML); (KNG)
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, United States of America
- * E-mail: (FML); (KNG)
| |
Collapse
|
220
|
Kitazawa M, Medeiros R, Laferla FM. Transgenic mouse models of Alzheimer disease: developing a better model as a tool for therapeutic interventions. Curr Pharm Des 2012; 18:1131-47. [PMID: 22288400 DOI: 10.2174/138161212799315786] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 12/19/2011] [Indexed: 12/13/2022]
Abstract
Alzheimer disease (AD) is the leading cause of dementia among elderly. Currently, no effective treatment is available for AD. Analysis of transgenic mouse models of AD has facilitated our understanding of disease mechanisms and provided valuable tools for evaluating potential therapeutic strategies. In this review, we will discuss the strengths and weaknesses of current mouse models of AD and the contribution towards understanding the pathological mechanisms and developing effective therapies.
Collapse
Affiliation(s)
- Masashi Kitazawa
- School of Natural Sciences, University of California, Merced, CA 95343, USA.
| | | | | |
Collapse
|
221
|
Consequences of inhibiting amyloid precursor protein processing enzymes on synaptic function and plasticity. Neural Plast 2012; 2012:272374. [PMID: 22792491 PMCID: PMC3390164 DOI: 10.1155/2012/272374] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 04/22/2012] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, one of whose major pathological hallmarks is the accumulation of amyloid plaques comprised of aggregated β-amyloid (Aβ) peptides. It is now recognized that soluble Aβ oligomers may lead to synaptic dysfunctions early in AD pathology preceding plaque deposition. Aβ is produced by a sequential cleavage of amyloid precursor protein (APP) by the activity of β- and γ-secretases, which have been identified as major candidate therapeutic targets of AD. This paper focuses on how Aβ alters synaptic function and the functional consequences of inhibiting the activity of the two secretases responsible for Aβ generation. Abnormalities in synaptic function resulting from the absence or inhibition of the Aβ-producing enzymes suggest that Aβ itself may have normal physiological functions which are disrupted by abnormal accumulation of Aβ during AD pathology. This interpretation suggests that AD therapeutics targeting the β- and γ-secretases should be developed to restore normal levels of Aβ or combined with measures to circumvent the associated synaptic dysfunction(s) in order to have minimal impact on normal synaptic function.
Collapse
|
222
|
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
|
223
|
Monenschein H, Horne DB, Bartberger MD, Hitchcock SA, Nguyen TT, Patel VF, Pennington LD, Zhong W. Structure guided P1′ modifications of HEA derived β-secretase inhibitors for the treatment of Alzheimer’s disease. Bioorg Med Chem Lett 2012; 22:3607-11. [DOI: 10.1016/j.bmcl.2012.04.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/05/2012] [Accepted: 04/10/2012] [Indexed: 11/29/2022]
|
224
|
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
|
225
|
Haass C, Kaether C, Thinakaran G, Sisodia S. Trafficking and proteolytic processing of APP. Cold Spring Harb Perspect Med 2012; 2:a006270. [PMID: 22553493 PMCID: PMC3331683 DOI: 10.1101/cshperspect.a006270] [Citation(s) in RCA: 769] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Accumulations of insoluble deposits of amyloid β-peptide are major pathological hallmarks of Alzheimer disease. Amyloid β-peptide is derived by sequential proteolytic processing from a large type I trans-membrane protein, the β-amyloid precursor protein. The proteolytic enzymes involved in its processing are named secretases. β- and γ-secretase liberate by sequential cleavage the neurotoxic amyloid β-peptide, whereas α-secretase prevents its generation by cleaving within the middle of the amyloid domain. In this chapter we describe the cell biological and biochemical characteristics of the three secretase activities involved in the proteolytic processing of the precursor protein. In addition we outline how the precursor protein maturates and traffics through the secretory pathway to reach the subcellular locations where the individual secretases are preferentially active. Furthermore, we illuminate how neuronal activity and mutations which cause familial Alzheimer disease affect amyloid β-peptide generation and therefore disease onset and progression.
Collapse
Affiliation(s)
- Christian Haass
- DZNE-German Center for Neurodegenerative Diseases, 80336 Munich, Germany; Adolf Butenandt-Institute, Biochemistry, Ludwig-Maximilians University, 80336 Munich, Germany.
| | | | | | | |
Collapse
|
226
|
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
|
227
|
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
|
228
|
Mao P, Manczak M, Calkins MJ, Truong Q, Reddy TP, Reddy AP, Shirendeb U, Lo HH, Rabinovitch PS, Reddy PH. Mitochondria-targeted catalase reduces abnormal APP processing, amyloid β production and BACE1 in a mouse model of Alzheimer's disease: implications for neuroprotection and lifespan extension. Hum Mol Genet 2012; 21:2973-90. [PMID: 22492996 DOI: 10.1093/hmg/dds128] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The purpose of this study was to investigate the protective effects of the mitochondria-targeted antioxidant catalase (MCAT) and lifespan extension in mice that express amyloid beta (Aβ). Using immunoblotting and immunostaining analyses, we measured the production of full-length amyloid precursor protein (APP), soluble APPα, C-terminal fragments CTF99 and CTF83, monomeric and oligomeric Aβ, Aβ deposits and beta site amyloid precursor protein cleaving enzyme 1 (BACE1), in different stages of disease progression in MCAT/AβPP and AβPP mice. Using quantitative reverse transcriptase polymerase chain reaction and immunostaining analyses, we studied the expression of catalase, BACE1, the Alzheimer's disease (AD) markers, synaptophysin, APP, neprilysin, insulin-degrading enzyme and transthyretin in MCAT, AβPP, MCAT/AβPP and wild-type (WT) mice. Using the high pressure liquid chromatography analysis of 8-hydroxy-2-deoxyguanosine, we measured oxidative DNA damage in the cerebral cortical tissues from MCAT, AβPP, MCAT/AβPP and WT mice. We found that the AβPP transgenic mice that carried the human MCAT gene lived 5 months longer than did the AβPP mice. We also found that the overexpression of MCAT in the brain sections from the MCAT/AβPP transgenic mice significantly correlated with a reduction in the levels of full-length APP, CTF99, BACE1, Aβ levels (40 and 42), Aβ deposits and oxidative DNA damage relative to the brain sections from the AβPP mice. Interestingly, we found significantly increased levels of soluble APPα and CTF83 in the MCAT/AβPP mice, relative to the AβPP mice. These data provide direct evidence that oxidative stress plays a primary role in AD etiopathology and that in MCAT mice express Aβ, MCAT prevents abnormal APP processing, reduces Aβ levels and enhances Aβ-degrading enzymes in mice at different ages, corresponding to different stages of disease progression. These findings indicate that mitochondria-targeted molecules may be an effective therapeutic approach to treat patients with AD.
Collapse
Affiliation(s)
- Peizhong Mao
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW185th Avenue, Beaverton, OR 97006, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
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
|
230
|
|
231
|
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
|
232
|
β-Secretases, Alzheimer's Disease, and Down Syndrome. Curr Gerontol Geriatr Res 2012; 2012:362839. [PMID: 22481915 PMCID: PMC3299320 DOI: 10.1155/2012/362839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/05/2011] [Indexed: 01/01/2023] Open
Abstract
Individuals with Down Syndrome (DS), or trisomy 21, develop Alzheimer's disease (AD) pathology by approximately 40 years of age. Chromosome 21 harbors several genes implicated in AD, including the amyloid precursor protein and one homologue of the β-site APP cleaving enzyme, BACE2. Processing of the amyloid precursor protein by β-secretase (BACE) is the rate-limiting step in the production of the pathogenic Aβ peptide. Increased amounts of APP in the DS brain result in increased amounts of Aβ and extracellular plaque formation beginning early in life. BACE dysregulation potentially represents an overlapping biological mechanism with sporadic AD and a common therapeutic target. As the lifespan for those with DS continues to increase, age-related concerns such as obesity, depression, and AD are of growing concern. The ability to prevent or delay the progression of neurodegenerative diseases will promote healthy aging and improve quality of life for those with DS.
Collapse
|
233
|
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
|
234
|
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
|
235
|
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
|
236
|
Abstract
β-Site APP-cleaving enzyme (BACE1) cleaves the amyloid precursor protein (APP) at the β-secretase site to initiate the production of Aβ peptides. These accumulate to form toxic oligomers and the amyloid plaques associated with Alzheimer's disease (AD). An increase of BACE1 levels in the brain of AD patients has been mostly attributed to alterations of its intracellular trafficking. Golgi-associated adaptor proteins, GGA sort BACE1 for export to the endosomal compartment, which is the major cellular site of BACE1 activity. BACE1 undergoes recycling between endosome, trans-Golgi network (TGN), and the plasma membrane, from where it is endocytosed and either further recycled or retrieved to the endosome. Phosphorylation of Ser498 facilitates BACE1 recognition by GGA1 for retrieval to the endosome. Ubiquitination of BACE1 C-terminal Lys501 signals GGA3 for exporting BACE1 to the lysosome for degradation. In addition, the retromer mediates the retrograde transport of BACE1 from endosome to TGN. Decreased levels of GGA proteins and increased levels of retromer-associated sortilin have been associated with AD. Both would promote the co-localization of BACE1 and the amyloid precursor protein in the TGN and endosomes. Decreased levels of GGA3 also impair BACE1 degradation. Further understanding of BACE1 trafficking and its regulation may offer new therapeutic approaches for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jiangli Tan
- Department of Pathology, and Mental Health Research Institute, The University of Melbourne, Parkville, Australia
| | | |
Collapse
|
237
|
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
|
238
|
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: 84] [Impact Index Per Article: 6.5] [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
|
239
|
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
|
240
|
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
|
241
|
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
|
242
|
Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu Y, Atwal J, Elliott JM, Prabhu S, Watts RJ, Dennis MS. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci Transl Med 2011; 3:84ra44. [PMID: 21613623 DOI: 10.1126/scitranslmed.3002230] [Citation(s) in RCA: 543] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Monoclonal antibodies have therapeutic potential for treating diseases of the central nervous system, but their accumulation in the brain is limited by the blood-brain barrier (BBB). Here, we show that reducing the affinity of an antibody for the transferrin receptor (TfR) enhances receptor-mediated transcytosis of the anti-TfR antibody across the BBB into the mouse brain where it reaches therapeutically relevant concentrations. Anti-TfR antibodies that bind with high affinity to TfR remain associated with the BBB, whereas lower-affinity anti-TfR antibody variants are released from the BBB into the brain and show a broad distribution 24 hours after dosing. We designed a bispecific antibody that binds with low affinity to TfR and with high affinity to the enzyme β-secretase (BACE1), which processes amyloid precursor protein into amyloid-β (Aβ) peptides including those associated with Alzheimer's disease. Compared to monospecific anti-BACE1 antibody, the bispecific antibody accumulated in the mouse brain and led to a greater reduction in brain Aβ after a single systemic dose. TfR-facilitated transcytosis of this bispecific antibody across the BBB may enhance its potency as an anti-BACE1 therapy for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Y Joy Yu
- Neurodegeneration Labs, Department of Neuroscience, Genentech Inc., South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
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]
|
244
|
Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci 2011; 89:939-45. [DOI: 10.1016/j.lfs.2011.09.023] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 08/16/2011] [Accepted: 09/28/2011] [Indexed: 01/02/2023]
|
245
|
Pharmacopore hypothesis generation of BACE-1 inhibitors and pharmacophore-driven identification of potent multi-target neuroprotective agents. Med Chem Res 2011. [DOI: 10.1007/s00044-011-9885-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
246
|
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
|
247
|
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
|
248
|
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
|
249
|
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]
|
250
|
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
|