1
|
Mishra AK, Srivastava A, Raj V, Saini V, Khan G, Singh H, Mishra A, Paliwal S. Recent Updates on Alzheimer's Disease: Pathogenesis, Pathophysiology, Molecular Approaches and Natural Bioactive Compounds Used in Contemporary Time to Alleviate Disease. Curr Alzheimer Res 2024; 21:538-556. [PMID: 39716788 DOI: 10.2174/0115672050361294241211071813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/25/2024]
Abstract
Alzheimer's disease (AD), characterised by gradual memory loss and neurodegeneration, is an important risk to global health. Despite the recent advances in the field of neuroscience, the complex biological mechanisms underlying the aetiology and pathology of AD have not been elucidated yet. The development of amyloid-beta plaques, hyperphosphorylation of tau protein, oxidative stress, and neuroinflammation have been identified as important components. The genesis of AD has been illuminated by advances in molecular techniques, which have shown the contributions of environmental, genetic, and epigenetic variables. Ongoing research is focused on the potential of bioactive compounds as therapeutic agents. Quercetin, epigallocatechin gallate, huperzine A, ginsenosides, omega-3 fatty acids, vitamin E, zinc, bacosides from brahmi, and withanolide A from ashwagandha are among the compounds that have demonstrated encouraging effects in modifying disease pathways. These bioactive substances demonstrate their potential for symptomatic relief by providing neuroprotective, antioxidant, anti-inflammatory, and cognitive-enhancing properties. The present review presents the recent findings on AD pathogenesis, molecular mechanisms, and the impact of natural compounds, offering a comprehensive perspective on current and emerging strategies for managing this debilitating condition.
Collapse
Affiliation(s)
| | | | - Varsha Raj
- Kharvel Subharti College of Pharmacy, Swami Vivekanand Subharti University, Meerut, 250005, India
| | - Vipin Saini
- MM College of Pharmacy, M.M. University, Ambala 133207, India
| | - Gyas Khan
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Saudia Arabia
| | - Harpreet Singh
- School of Pharmaceutical Sciences, IFTM University, Moradabad 244102, India
| | - Amrita Mishra
- Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| |
Collapse
|
2
|
Embedded in the Membrane: How Lipids Confer Activity and Specificity to Intramembrane Proteases. J Membr Biol 2017; 251:369-378. [PMID: 29260282 DOI: 10.1007/s00232-017-0008-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/12/2017] [Indexed: 10/18/2022]
Abstract
Proteases, sharp yet unforgivable tools of every cell, require tight regulation to ensure specific non-aberrant cleavages. The relatively recent discovered class of intramembrane proteases has gained increasing interest due to their involvement in important signaling pathways linking them to diseases including Alzheimer's disease and cancer. Despite tremendous efforts, their regulatory mechanisms have only started to unravel. There is evidence that the membrane composition itself can regulate intramembrane protease activity and specificity. In this review, we highlight the work on γ-secretase and rhomboid proteases and summarize several studies as to how different lipids impact on enzymatic activity.
Collapse
|
3
|
Affiliation(s)
- Rodrigo Aguayo-Ortiz
- Departamento de Fisicoquímica; Universidad Nacional Autónoma de México; Ciudad de México 04510 México
| | - Laura Dominguez
- Departamento de Fisicoquímica; Universidad Nacional Autónoma de México; Ciudad de México 04510 México
| |
Collapse
|
4
|
Crenshaw E, Leung BP, Kwok CK, Sharoni M, Olson K, Sebastian NP, Ansaloni S, Schweitzer-Stenner R, Akins MR, Bevilacqua PC, Saunders AJ. Amyloid Precursor Protein Translation Is Regulated by a 3'UTR Guanine Quadruplex. PLoS One 2015; 10:e0143160. [PMID: 26618502 PMCID: PMC4664259 DOI: 10.1371/journal.pone.0143160] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022] Open
Abstract
A central event in Alzheimer’s disease is the accumulation of amyloid β (Aβ) peptides generated by the proteolytic cleavage of the amyloid precursor protein (APP). APP overexpression leads to increased Aβ generation and Alzheimer’s disease in humans and altered neuronal migration and increased long term depression in mice. Conversely, reduction of APP expression results in decreased Aβ levels in mice as well as impaired learning and memory and decreased numbers of dendritic spines. Together these findings indicate that therapeutic interventions that aim to restore APP and Aβ levels must do so within an ideal range. To better understand the effects of modulating APP levels, we explored the mechanisms regulating APP expression focusing on post-transcriptional regulation. Such regulation can be mediated by RNA regulatory elements such as guanine quadruplexes (G-quadruplexes), non-canonical structured RNA motifs that affect RNA stability and translation. Via a bioinformatics approach, we identified a candidate G-quadruplex within the APP mRNA in its 3’UTR (untranslated region) at residues 3008–3027 (NM_201414.2). This sequence exhibited characteristics of a parallel G-quadruplex structure as revealed by circular dichroism spectrophotometry. Further, as with other G-quadruplexes, the formation of this structure was dependent on the presence of potassium ions. This G-quadruplex has no apparent role in regulating transcription or mRNA stability as wild type and mutant constructs exhibited equivalent mRNA levels as determined by real time PCR. Instead, we demonstrate that this G-quadruplex negatively regulates APP protein expression using dual luciferase reporter and Western blot analysis. Taken together, our studies reveal post-transcriptional regulation by a 3’UTR G-quadruplex as a novel mechanism regulating APP expression.
Collapse
Affiliation(s)
- Ezekiel Crenshaw
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Brian P. Leung
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
- Department of Chemistry, Drexel University, Philadelphia, PA, United States of America
| | - Chun Kit Kwok
- Department of Chemistry, Pennsylvania State University, University Park, PA, United States of America
- Department of Biochemistry & Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA, United States of America
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Michal Sharoni
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Kalee Olson
- Department of Chemistry, Pennsylvania State University, University Park, PA, United States of America
| | - Neeraj P. Sebastian
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Sara Ansaloni
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | | | - Michael R. Akins
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Philip C. Bevilacqua
- Department of Chemistry, Pennsylvania State University, University Park, PA, United States of America
- Department of Biochemistry & Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA, United States of America
| | - Aleister J. Saunders
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
5
|
Osolodkin DI, Radchenko EV, Orlov AA, Voronkov AE, Palyulin VA, Zefirov NS. Progress in visual representations of chemical space. Expert Opin Drug Discov 2015; 10:959-73. [DOI: 10.1517/17460441.2015.1060216] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
6
|
Zhang H, Gao Y, Qiao P, Zhao F, Yan Y. Fenofibrate reduces amyloidogenic processing of APP in APP/PS1 transgenic mice via PPAR‐α/PI3‐K pathway. Int J Dev Neurosci 2014; 38:223-31. [DOI: 10.1016/j.ijdevneu.2014.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/12/2014] [Accepted: 10/21/2014] [Indexed: 11/26/2022] Open
Affiliation(s)
- Hua Zhang
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Ying Gao
- Special WardsThe Affiliated Children's Hospital of Chongqing Medical UniversityChongqing400014China
| | - Pei‐feng Qiao
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Feng‐li Zhao
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Yong Yan
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| |
Collapse
|
7
|
Mishima K, Kaneko H, Funatsu K. Development of a New De Novo Design Algorithm for Exploring Chemical Space. Mol Inform 2014; 33:779-89. [PMID: 27485424 DOI: 10.1002/minf.201400056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 07/29/2014] [Indexed: 01/10/2023]
Abstract
In the first stage of development of new drugs, various lead compounds with high activity are required. To design such compounds, we focus on chemical space defined by structural descriptors. New compounds close to areas where highly active compounds exist will show the same degree of activity. We have developed a new de novo design system to search a target area in chemical space. First, highly active compounds are manually selected as initial seeds. Then, the seeds are entered into our system, and structures slightly different from the seeds are generated and pooled. Next, seeds are selected from the new structure pool based on the distance from target coordinates on the map. To test the algorithm, we used two datasets of ligand binding affinity and showed that the proposed generator could produce diverse virtual compounds that had high activity in docking simulations.
Collapse
Affiliation(s)
- Kazuaki Mishima
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan tel:(+81) 03-5841-7751
| | - Hiromasa Kaneko
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan tel:(+81) 03-5841-7751
| | - Kimito Funatsu
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan tel:(+81) 03-5841-7751.
| |
Collapse
|
8
|
Branca C, Sarnico I, Ruotolo R, Lanzillotta A, Viscomi AR, Benarese M, Porrini V, Lorenzini L, Calzà L, Imbimbo BP, Ottonello S, Pizzi M. Pharmacological targeting of the β-amyloid precursor protein intracellular domain. Sci Rep 2014; 4:4618. [PMID: 24714650 PMCID: PMC3980230 DOI: 10.1038/srep04618] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 03/18/2014] [Indexed: 11/09/2022] Open
Abstract
Amyloid precursor protein (APP) intracellular domain (AICD) is a product of APP processing with transcriptional modulation activity, whose overexpression causes various Alzheimer's disease (AD)-related dysfunctions. Here we report that 1-(3',4'-dichloro-2-fluoro[1,1'-biphenyl]-4-yl)-cyclopropanecarboxylic acid) (CHF5074), a compound that favorably affects neurodegeneration, neuroinflammation and memory deficit in transgenic mouse models of AD, interacts with the AICD and impairs its nuclear activity. In neuroglioma-APPswe cells, CHF5074 shifted APP cleavage from Aβ42 to the less toxic Aβ38 peptide without affecting APP-C-terminal fragment, nor APP levels. As revealed by photoaffinity labeling, CHF5074 does not interact with γ-secretase, but binds to the AICD and lowers its nuclear translocation. In vivo treatment with CHF5074 reduced AICD occupancy as well as histone H3 acetylation levels and transcriptional output of the AICD-target gene KAI1. The data provide new mechanistic insights on this compound, which is under clinical investigation for AD treatment/prevention, as well as on the contribution of the AICD to AD pathology.
Collapse
Affiliation(s)
- Caterina Branca
- 1] Department of Molecular & Translational Medicine and National Institute of Neuroscience, School of Medicine, University of Brescia, Italy [2]
| | - Ilenia Sarnico
- 1] Department of Molecular & Translational Medicine and National Institute of Neuroscience, School of Medicine, University of Brescia, Italy [2]
| | - Roberta Ruotolo
- Laboratory of Functional Genomics and Protein Engineering, Biochemistry and Molecular Biology Unit, Department of Life Sciences, University of Parma, Italy
| | - Annamaria Lanzillotta
- Department of Molecular & Translational Medicine and National Institute of Neuroscience, School of Medicine, University of Brescia, Italy
| | - Arturo Roberto Viscomi
- 1] Laboratory of Functional Genomics and Protein Engineering, Biochemistry and Molecular Biology Unit, Department of Life Sciences, University of Parma, Italy [2]
| | - Marina Benarese
- Department of Molecular & Translational Medicine and National Institute of Neuroscience, School of Medicine, University of Brescia, Italy
| | - Vanessa Porrini
- 1] Department of Molecular & Translational Medicine and National Institute of Neuroscience, School of Medicine, University of Brescia, Italy [2] IRCCS, San Camillo Hospital, Venice, Italy
| | - Luca Lorenzini
- Department of Veterinary Medicine and Health Science, University of Bologna, Italy
| | - Laura Calzà
- Department of Veterinary Medicine and Health Science, University of Bologna, Italy
| | | | - Simone Ottonello
- Laboratory of Functional Genomics and Protein Engineering, Biochemistry and Molecular Biology Unit, Department of Life Sciences, University of Parma, Italy
| | - Marina Pizzi
- 1] Department of Molecular & Translational Medicine and National Institute of Neuroscience, School of Medicine, University of Brescia, Italy [2] IRCCS, San Camillo Hospital, Venice, Italy
| |
Collapse
|
9
|
Dimitrov M, Alattia JR, Lemmin T, Lehal R, Fligier A, Houacine J, Hussain I, Radtke F, Dal Peraro M, Beher D, Fraering PC. Alzheimer's disease mutations in APP but not γ-secretase modulators affect epsilon-cleavage-dependent AICD production. Nat Commun 2014; 4:2246. [PMID: 23907250 DOI: 10.1038/ncomms3246] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 07/03/2013] [Indexed: 12/29/2022] Open
Abstract
Pathological amino-acid substitutions in the amyloid precursor protein (APP) and chemical γ-secretase modulators affect the processing of APP by the γ-secretase complex and the production of the amyloid-beta peptide Aβ42, the accumulation of which is considered causative of Alzheimer's disease. Here we demonstrate that mutations in the transmembrane domain of APP causing aggressive early-onset familial Alzheimer's disease affect both γ- and ε-cleavage sites, by raising the Aβ42/40 ratio and inhibiting the production of AICD50-99, one of the two physiological APP intracellular domains (ICDs). This is in sharp contrast to γ-secretase modulators, which shift Aβ42 production towards the shorter Aβ38, but unequivocally spare the ε-site and APP- and Notch-ICDs production. Molecular simulations suggest that familial Alzheimer's disease mutations modulate the flexibility of the APP transmembrane domain and the presentation of its γ-site, modifying at the same time, the solvation of the ε-site.
Collapse
Affiliation(s)
- Mitko Dimitrov
- Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Martell J, Weerapana E. Applications of copper-catalyzed click chemistry in activity-based protein profiling. Molecules 2014; 19:1378-93. [PMID: 24473203 PMCID: PMC6270908 DOI: 10.3390/molecules19021378] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/09/2014] [Accepted: 01/17/2014] [Indexed: 12/24/2022] Open
Abstract
Activity-based protein profiling (ABPP) is a chemical proteomic technique that enables the interrogation of protein activity directly within complex proteomes. Given the dominant role of posttranslational modifications in regulating protein function in vivo, ABPP provides a direct readout of activity that is not attained through traditional proteomic methods. ABPP relies on the design of covalent binding probes that either target a specific enzyme or a class of enzymes with related function. These covalent warheads are coupled to either fluorophores or biotin groups for visualization and enrichment of these active proteins. The advent of bioorthogonal chemistries, in particular, the copper (I)-catalyzed azide-alkyne cycloaddition (CuAAC), has benefitted the field of ABPP by achieving the following: (1) replacing bulky reporter groups with smaller alkyne or azide groups to promote cell permeability; (2) adding modularity to the system such that a single probe can be diversified with a variety of reporter groups without the need to develop new synthetic routes; and (3) enabling the conjugation of complex linkers to facilitate quantitative proteomic analyses. Here, we summarize recent examples of CuAAC in ABPP that serve to illustrate the contribution of bioorthogonal chemistry to advancing discoveries in this field.
Collapse
Affiliation(s)
- Julianne Martell
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA.
| | | |
Collapse
|
11
|
Song Y, Hustedt EJ, Brandon S, Sanders CR. Competition between homodimerization and cholesterol binding to the C99 domain of the amyloid precursor protein. Biochemistry 2013; 52:5051-64. [PMID: 23865807 DOI: 10.1021/bi400735x] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The 99-residue transmembrane C-terminal domain (C99, also known as β-CTF) of the amyloid precursor protein (APP) is the product of the β-secretase cleavage of the full-length APP and is the substrate for γ-secretase cleavage. The latter cleavage releases the amyloid-β polypeptides that are closely associated with Alzheimer's disease. C99 is thought to form homodimers; however, the free energy in favor of dimerization has not previously been quantitated. It was also recently documented that cholesterol forms a 1:1 complex with monomeric C99 in bicelles. Here, the affinities for both homodimerization and cholesterol binding to C99 were measured in bilayered lipid vesicles using both electron paramagnetic resonance (EPR) and Förster resonance energy transfer (FRET) methods. Homodimerization and cholesterol binding were seen to be competitive processes that center on the transmembrane G₇₀₀XXXG₇₀₄XXXG₇₀₈ glycine-zipper motif and adjacent Gly709. On one hand, the observed Kd for cholesterol binding (Kd = 2.7 ± 0.3 mol %) is on the low end of the physiological cholesterol concentration range in mammalian cell membranes. On the other hand, the observed K(d) for homodimerization (K(d) = 0.47 ± 0.15 mol %) likely exceeds the physiological concentration range for C99. These results suggest that the 1:1 cholesterol/C99 complex will be more highly populated than C99 homodimers under most physiological conditions. These observations are of relevance for understanding the γ-secretase cleavage of C99.
Collapse
Affiliation(s)
- Yuanli Song
- Department of Biochemistry and Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | | | | | | |
Collapse
|
12
|
Pozdnyakov N, Murrey HE, Crump CJ, Pettersson M, Ballard TE, Am Ende CW, Ahn K, Li YM, Bales KR, Johnson DS. γ-Secretase modulator (GSM) photoaffinity probes reveal distinct allosteric binding sites on presenilin. J Biol Chem 2013; 288:9710-9720. [PMID: 23396974 DOI: 10.1074/jbc.m112.398602] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
γ-Secretase is an intramembrane aspartyl protease that cleaves the amyloid precursor protein to produce neurotoxic β-amyloid peptides (i.e. Aβ42) that have been implicated in the pathogenesis of Alzheimer disease. Small molecule γ-secretase modulators (GSMs) have emerged as potential disease-modifying treatments for Alzheimer disease because they reduce the formation of Aβ42 while not blocking the processing of γ-secretase substrates. We developed clickable GSM photoaffinity probes with the goal of identifying the target of various classes of GSMs and to better understand their mechanism of action. Here, we demonstrate that the photoaffinity probe E2012-BPyne specifically labels the N-terminal fragment of presenilin-1 (PS1-NTF) in cell membranes as well as in live cells and primary neuronal cultures. The labeling is competed in the presence of the parent imidazole GSM E2012, but not with acid GSM-1, allosteric GSI BMS-708163, or substrate docking site peptide inhibitor pep11, providing evidence that these compounds have distinct binding sites. Surprisingly, we found that the cross-linking of E2012-BPyne to PS1-NTF is significantly enhanced in the presence of the active site-directed GSI L-685,458 (L458). In contrast, L458 does not affect the labeling of the acid GSM photoprobe GSM-5. We also observed that E2012-BPyne specifically labels PS1-NTF (active γ-secretase) but not full-length PS1 (inactive γ-secretase) in ANP.24 cells. Taken together, our results support the hypothesis that multiple binding sites within the γ-secretase complex exist, each of which may contribute to different modes of modulatory action. Furthermore, the enhancement of PS1-NTF labeling by E2012-BPyne in the presence of L458 suggests a degree of cooperativity between the active site of γ-secretase and the modulatory binding site of certain GSMs.
Collapse
Affiliation(s)
- Nikolay Pozdnyakov
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Heather E Murrey
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Christina J Crump
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; Department of Pharmacology, Weill Graduate School of Medical Science of Cornell University, New York, New York 10021
| | - Martin Pettersson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - T Eric Ballard
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | | | - Kwangwook Ahn
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; Department of Pharmacology, Weill Graduate School of Medical Science of Cornell University, New York, New York 10021
| | - Yue-Ming Li
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; Department of Pharmacology, Weill Graduate School of Medical Science of Cornell University, New York, New York 10021
| | - Kelly R Bales
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139.
| | - Douglas S Johnson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139.
| |
Collapse
|
13
|
γ-Secretase Modulators: Can We Combine Potency with Safety? Int J Alzheimers Dis 2012; 2012:295207. [PMID: 23365783 PMCID: PMC3534213 DOI: 10.1155/2012/295207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/08/2012] [Indexed: 01/23/2023] Open
Abstract
γ-Secretase modulation has been proposed as a potential disease modifying anti-Alzheimer's approach. γ-Secretase modulators (GSMs) cause a product shift from the longer amyloid-beta (Aβ) peptide isoforms to shorter, more soluble, and less amyloidogenic isoforms, without inhibiting APP or Notch proteolytic processing. As such, modulating γ-secretase may avoid some of the adverse effects observed with γ-secretase inhibitors. Since the termination of the GSM tarenfurbil in 2008 due to negative phase III trial results, a considerable progress has been made towards more potent and better brain penetrable compounds. However, an analysis of their lipophilic efficiency indices indicates that their increased potency can be largely attributed to their increased lipophilicity. The need for early and chronic dosing with GSMs will require high-safety margins. This will be a challenge to achieve with the current, highly lipophilic GSMs. We will demonstrate that by focusing on the drug-like properties of GSMs, a combination of high in vitro potency and reduced lipophilicity can be achieved and does result in better tolerated compounds. The next hurdle will be to translate this knowledge into GSMs which are highly efficacious and safe in vivo.
Collapse
|
14
|
Zettl H, Ness J, Hähnke V, Beher D, Jumpertz T, Saric A, Baumann K, Pietrzik CU, Bulic B, Schneider G, Weggen S. Discovery of γ-secretase modulators with a novel activity profile by text-based virtual screening. ACS Chem Biol 2012; 7:1488-95. [PMID: 22725102 DOI: 10.1021/cb3001952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We present an integrated approach to identify and optimize a novel class of γ-secretase modulators (GSMs) with a unique pharmacological profile. Our strategy included (i) virtual screening through application of a recently developed protocol (PhAST), (ii) synthetic chemistry to discover structure-activity relationships, and (iii) detailed in vitro pharmacological characterization. GSMs are promising agents for treatment or prevention of Alzheimer's disease. They modulate the γ-secretase product spectrum (i.e., amyloid-β (Aβ) peptides of different length) and induce a shift from toxic Aβ42 to shorter Aβ species such as Aβ38 with no or minimal effect on the overall rate of γ-secretase cleavage. We describe the identification of a series of 4-hydroxypyridin-2-one derivatives, which display a novel type of γ-secretase modulation with equipotent inhibition of Aβ42 and Aβ38 peptide species.
Collapse
Affiliation(s)
- Heiko Zettl
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Julia Ness
- Department of Neuropathology, Heinrich-Heine-University, D-40225 Duesseldorf, Germany
| | - Volker Hähnke
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Dirk Beher
- Global Research & Early Development, Merck Serono S. A., CH-1202 Geneva, Switzerland
| | - Thorsten Jumpertz
- Department of Neuropathology, Heinrich-Heine-University, D-40225 Duesseldorf, Germany
| | - Arman Saric
- Global Research & Early Development, Merck Serono S. A., CH-1202 Geneva, Switzerland
| | - Karlheinz Baumann
- F. Hoffmann-La Roche AG, pRED, Pharma Research & Early Development, DTA CNS, CH-4070 Basel, Switzerland
| | - Claus U. Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, D-55128 Mainz, Germany
| | - Bruno Bulic
- Research Group Chemical Biology of Neurodegenerative Diseases, Center of Advanced European Studies and Research, D-53175 Bonn, Germany
| | - Gisbert Schneider
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, D-40225 Duesseldorf, Germany
| |
Collapse
|
15
|
Bischoff F, Berthelot D, De Cleyn M, Macdonald G, Minne G, Oehlrich D, Pieters S, Surkyn M, Trabanco AA, Tresadern G, Van Brandt S, Velter I, Zaja M, Borghys H, Masungi C, Mercken M, Gijsen HJM. Design and Synthesis of a Novel Series of Bicyclic Heterocycles As Potent γ-Secretase Modulators. J Med Chem 2012; 55:9089-106. [DOI: 10.1021/jm201710f] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Francois Bischoff
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Didier Berthelot
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Michel De Cleyn
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Gregor Macdonald
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Garrett Minne
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Daniel Oehlrich
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Serge Pieters
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Michel Surkyn
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Andrés A. Trabanco
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Gary Tresadern
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Sven Van Brandt
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Ingrid Velter
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Mirko Zaja
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Herman Borghys
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Chantal Masungi
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Marc Mercken
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | - Harrie J. M. Gijsen
- Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| |
Collapse
|
16
|
Pettersson M, Johnson DS, Subramanyam C, Bales KR, am Ende CW, Fish BA, Green ME, Kauffman GW, Lira R, Mullins PB, Navaratnam T, Sakya SM, Stiff CM, Tran TP, Vetelino BC, Xie L, Zhang L, Pustilnik LR, Wood KM, O'Donnell CJ. Design and synthesis of dihydrobenzofuran amides as orally bioavailable, centrally active γ-secretase modulators. Bioorg Med Chem Lett 2012; 22:2906-11. [PMID: 22429469 DOI: 10.1016/j.bmcl.2012.02.059] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 12/12/2022]
Abstract
We report the discovery and optimization of a novel series of dihydrobenzofuran amides as γ-secretase modulators (GSMs). Strategies for aligning in vitro potency with drug-like physicochemical properties and good microsomal stability while avoiding P-gp mediated efflux are discussed. Lead compounds such as 35 and 43 have moderate to good in vitro potency and excellent selectivity against Notch. Good oral bioavailability was achieved as well as robust brain Aβ42 lowering activity at 100 mg/kg po dose.
Collapse
Affiliation(s)
- Martin Pettersson
- Neuroscience Medicinal Chemistry, Pfizer Worldwide Research and Development, Groton, CT 06340, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
DOGS: reaction-driven de novo design of bioactive compounds. PLoS Comput Biol 2012; 8:e1002380. [PMID: 22359493 PMCID: PMC3280956 DOI: 10.1371/journal.pcbi.1002380] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 12/21/2011] [Indexed: 11/19/2022] Open
Abstract
We present a computational method for the reaction-based de novo design of drug-like molecules. The software DOGS (Design of Genuine Structures) features a ligand-based strategy for automated ‘in silico’ assembly of potentially novel bioactive compounds. The quality of the designed compounds is assessed by a graph kernel method measuring their similarity to known bioactive reference ligands in terms of structural and pharmacophoric features. We implemented a deterministic compound construction procedure that explicitly considers compound synthesizability, based on a compilation of 25'144 readily available synthetic building blocks and 58 established reaction principles. This enables the software to suggest a synthesis route for each designed compound. Two prospective case studies are presented together with details on the algorithm and its implementation. De novo designed ligand candidates for the human histamine H4 receptor and γ-secretase were synthesized as suggested by the software. The computational approach proved to be suitable for scaffold-hopping from known ligands to novel chemotypes, and for generating bioactive molecules with drug-like properties. The computer program DOGS aims at the automated generation of new bioactive compounds. Only a single known reference compound is required to have the computer come up with suggestions for potentially isofunctional molecules. A specific feature of the algorithm is its capability to propose a synthesis plan for each designed compound, based on a large set of readily available molecular building blocks and established reaction protocols. The de novo design software provides rapid access to tool compounds and starting points for the development of a lead candidate structure. The manuscript gives a detailed description of the algorithm. Theoretical analysis and prospective case studies demonstrate its ability to propose bioactive, plausible and chemically accessible compounds.
Collapse
|
18
|
Barrett PJ, Sanders CR, Kaufman SA, Michelsen K, Jordan JB. NSAID-based γ-secretase modulators do not bind to the amyloid-β polypeptide. Biochemistry 2011; 50:10328-42. [PMID: 21995415 DOI: 10.1021/bi201371j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
γ-Secretase modulators (GSMs) have received much attention as potential therapeutic agents for Alzheimer's disease (AD). GSMs increase the ratio between short and long forms of the amyloid-β (Aβ) polypeptides produced by γ-secretase and thereby decrease the amount of the toxic amyloid species. However, the mechanism of action of these agents is still poorly understood. One recent paper [Richter et al. (2010) Proc. Natl. Acad. Sci. U. S. A.107, 14597-14602] presented data that were interpreted to support direct binding of the GSM sulindac sulfide to Aβ(42), supporting the notion that GSM action is linked to direct binding of these compounds to the Aβ domain of its immediate precursor, the 99-residue C-terminal domain of the amyloid precursor protein (C99, also known as the β-CTF). Here, contrasting results are presented that indicate there is no interaction between monomeric sulindac sulfide and monomeric forms of Aβ42. Instead, it was observed that sulindac sulfide is itself prone to form aggregates that can bind nonspecifically to Aβ42 and trigger its aggregation. This observation, combined with data from previous work [Beel et al. (2009) Biochemistry48, 11837-11839], suggests both that the poor behavior of some NSAID-based GSMs in solution may obscure results of binding assays and that NSAID-based GSMs do not function by directly targeting C99. It was also observed that another GSM, flurbiprofen, fails to bind to monomeric Aβ42 or to C99 reconstituted into bilayered lipid vesicles. These results disfavor the hypothesis that these NSAID-based GSMs exert their modulatory effect by directly targeting a site located in the Aβ42 domain of free C99.
Collapse
Affiliation(s)
- Paul J Barrett
- Department of Biochemistry and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232-8725, United States
| | | | | | | | | |
Collapse
|
19
|
Sagi SA, Lessard CB, Winden KD, Maruyama H, Koo JC, Weggen S, Kukar TL, Golde TE, Koo EH. Substrate sequence influences γ-secretase modulator activity, role of the transmembrane domain of the amyloid precursor protein. J Biol Chem 2011; 286:39794-803. [PMID: 21868380 DOI: 10.1074/jbc.m111.277228] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A subset of non-steroidal anti-inflammatory drugs modulates the γ cleavage site in the amyloid precursor protein (APP) to selectively reduce production of Aβ42. It is unclear precisely how these γ-secretase modulators (GSMs) act to preferentially spare Aβ40 production as well as Notch processing and signaling. In an effort to determine the substrate requirements in NSAID/GSM activity, we determined the effects of sulindac sulfide and flurbiprofen on γ-cleavage of artificial constructs containing several γ-secretase substrates. Using FLAG-tagged constructs that expressed extracellularly truncated APP, Notch-1, or CD44, we found that these substrates have different sensitivities to sulindac sulfide. γ-Secretase cleavage of APP was altered by sulindac sulfide, but CD44 and Notch-1 were either insensitive or only minimally altered by this compound. Using chimeric APP constructs, we observed that the transmembrane domain (TMD) of APP played a pivotal role in determining drug sensitivity. Substituting the APP TMD with that of APLP2 retained the sensitivity to γ-cleavage modulation, but replacing TMDs from Notch-1 or ErbB4 rendered the resultant molecules insensitive to drug treatment. Specifically, the GXXXG motif within APP appeared to be critical to GSM activity. Consequently, the modulatory effects on γ-cleavage appears to be substrate-dependent. We hypothesize that the substrate present in the γ-secretase complex influences the conformation of the complex so that the binding site of GSMs is either stabilized or less favorable to influence the cleavage of the respective substrates.
Collapse
Affiliation(s)
- Sarah A Sagi
- Department of Neurosciences, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hieke M, Ness J, Steri R, Greiner C, Werz O, Schubert-Zsilavecz M, Weggen S, Zettl H. SAR studies of acidic dual γ-secretase/PPARγ modulators. Bioorg Med Chem 2011; 19:5372-82. [PMID: 21873070 DOI: 10.1016/j.bmc.2011.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/25/2011] [Accepted: 08/02/2011] [Indexed: 11/25/2022]
Abstract
A novel set of dual γ-secretase/PPARγ modulators characterized by a 2-benzyl hexanoic acid scaffold is presented. Synthetic efforts were focused on the variation of the substitution pattern of the central benzene. Finally, we obtained a new class of 2,5-disubstituted 2-benzylidene hexanoic acid derivatives, which act as dual γ-secretase/PPARγ modulators in the low micromolar range. We have explored broad SAR and successfully improved the dual pharmacological activity and the selectivity profile against potential off-targets such as NOTCH and COX. Compound 17 showed an IC(50) Aβ42=2.4 μM and an EC(50) PPARγ=7.2 μM and could be a valuable tool to further evaluate the concept of dual γ-secretase/PPARγ modulators in animal models of Alzheimer's disease.
Collapse
Affiliation(s)
- Martina Hieke
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Fischer C, Zultanski SL, Zhou H, Methot JL, Brown WC, Mampreian DM, Schell AJ, Shah S, Nuthall H, Hughes BL, Smotrov N, Kenific CM, Cruz JC, Walker D, Bouthillette M, Nikov GN, Savage DF, Jeliazkova-Mecheva VV, Diaz D, Szewczak AA, Bays N, Middleton RE, Munoz B, Shearman MS. Triazoles as γ-secretase modulators. Bioorg Med Chem Lett 2011; 21:4083-7. [DOI: 10.1016/j.bmcl.2011.04.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 04/18/2011] [Accepted: 04/21/2011] [Indexed: 11/26/2022]
|
22
|
Pettersson M, Kauffman GW, am Ende CW, Patel NC, Stiff C, Tran TP, Johnson DS. Novel γ-secretase modulators: a review of patents from 2008 to 2010. Expert Opin Ther Pat 2011; 21:205-26. [PMID: 21231889 DOI: 10.1517/13543776.2011.547479] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The amyloid precursor protein is first cleaved by β-secretase to generate a 99-residue membrane-bound CTF (C99 or β-CTF), which is subsequently cleaved by γ-secretase to generate amyloid β (Aβ) peptides and the APP intracellular domain. The amyloidogenic Aβ42 has attracted considerable attention because it is thought to be the most pathogenic species associated with Alzheimer's disease progression. New classes of compounds, called γ-secretase modulators (GSMs), have been shown to selectively lower Aβ42 production without shutting down key γ-secretase-dependent signaling pathways. This has become an important therapeutic strategy aimed at modulating Aβ production. AREAS COVERED The progress on the clinical development of γ-secretase inhibitors is briefly covered in this review, followed by a discussion of the potential differentiating attributes of GSMs. Then, the patent literature covering novel GSMs is reviewed, focusing on patents from 2008 to 2010. EXPERT OPINION Much progress has been made in the past 2 years on developing GSMs with improved potency for lowering the production of Aβ42. However, many of these chemotypes are in a challenging chemical space and generally possess higher lipophilicity than most CNS drugs. It will be important to gain a better understanding of the specific target(s) that these GSMs interact with in order to facilitate future drug design efforts.
Collapse
Affiliation(s)
- Martin Pettersson
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern Point Road, Groton, CT 06340, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Hahn S, Brüning T, Ness J, Czirr E, Baches S, Gijsen H, Korth C, Pietrzik CU, Bulic B, Weggen S. Presenilin-1 but not amyloid precursor protein mutations present in mouse models of Alzheimer's disease attenuate the response of cultured cells to γ-secretase modulators regardless of their potency and structure. J Neurochem 2010; 116:385-95. [PMID: 21091478 DOI: 10.1111/j.1471-4159.2010.07118.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
γ-Secretase modulators (GSMs) inhibit the generation of amyloidogenic Aβ42 peptides and are promising agents for treatment or prevention of Alzheimer's disease (AD). Recently, a second generation of GSMs with favorable pharmacological properties has emerged, but preclinical studies to assess their efficacy in vivo are lacking. Such studies rely on transgenic mouse models that express amyloid precursor protein (APP) and presenilin (PSEN) mutations associated with early-onset familial AD. Previously, we have shown that certain PSEN1 mutations attenuated the response of cultured cells to GSMs and potentially confound in vivo studies in AD mouse models. However, different combinations of familial AD mutations might have synergistic or opposing effects, and we have now systematically determined the response of APP and PSEN1 mutations present in current AD models. Using a potent acidic GSM, we found that APP mutations, either single mutations or in combination, did not affect the potency of GSMs. In contrast, all PSEN1 mutations that have been used to accelerate pathological changes in AD models strongly attenuated the Aβ42-lowering activity of GSMs with two exceptions (M146L, A246E). Similar results were obtained with potent non-acidic GSMs indicating that the attenuating effect of PSEN1 mutations cannot simply be overcome by increased potency or structural changes. Notably, two non-acidic compounds fully compensated the attenuating effect of the PSEN1-G384A mutation. Taken together, our findings indicate that most AD models with rapid pathology and advanced phenotypes are unsuitable for preclinical GSM studies. However, we also provide evidence that additional compound screens could discover GSMs that are able to break the attenuating effects of PSEN mutations.
Collapse
Affiliation(s)
- Stefanie Hahn
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|